Methods for assessing risk of developing a viral disease using a genetic test

Abstract
This document provides methods and materials related to treating a disease. For example, this document provides methods for treating a subject's disease based on identifying the risk of progressive multifocal leukoencephalopathy PML using a genetic test.
Description
REFERENCE TO A SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on Mar. 15, 2023, is named 56969_702_404_SL.xml and is 80,955,165 bytes in size.


BACKGROUND OF THE DISCLOSURE

Progressive multifocal leukoencephalopathy (PML) is a rare and potentially fatal opportunistic infection of the central nervous system that is caused by a ubiquitous polyomavirus, the JC virus (JCV). While JCV is present at very high rates in the general population, PML remains a rare disorder, albeit an important one because of the poor survival and the severe neurological sequelae, and the recently demonstrated association with a variety of useful therapies, for example, natalizumab in multiple sclerosis (MS). A number of risk factors for PML have been described but these are better viewed as necessary but not sufficient. While these risk factors are highly relevant, they do not, on their own, predict who will develop PML, since the vast majority of individuals with these risk factors will not develop the disorder. Other factors need to be considered and there is growing evidence for the role of host genetic factors in susceptibility to PML.


The ability to more accurately predict who is at risk of developing PML will be of enormous benefit in the context of drug treatment with compounds that are highly effective in their disease context (natalizumab in MS, for example) but carry a small risk of a devastating disorder. There is a need to develop a companion diagnostic testing, in order to effectively exclude those that were at risk of PML, in the process reassuring those with negative tests about their dramatically reduced risk of developing PML.


INCORPORATION BY REFERENCE

All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. In the event of a conflict between a term herein and a term incorporated by reference, the term herein controls.





BRIEF DESCRIPTION OF THE DRAWINGS

The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the disclosure are utilized, and the accompanying drawings.



FIG. 1 represents an example of a gene (PRKCB) impacted by germline and acquired CNVs.



FIG. 2 represents an example of genes (TNFRSF13C and CENPM) impacted by acquired CNVs.



FIG. 3 represents an example of a gene (PKHD1) impacted by germline and acquired CNVs.



FIG. 4 represents an example of a gene (BMPR2) impacted by a recurrent CNV (homozygous and heterozygous losses).



FIG. 5 represents an example of a gene (COMMD6) impacted by a recurrent CNV (e.g., homozygous duplication).



FIG. 6 represents an example of genes (KCTD7, RABGEF1) directly and potentially impacted by a recurrent CNV (e.g., homozygous duplication).



FIG. 7 represents an example of a gene (FPR2) impacted by a recurrent CNV (e.g., homozygous duplication).



FIG. 8 represents an example of a gene (PIK3CD) impacted by a CNV (e.g., homozygous loss).



FIG. 9 represents an example of a gene (CD180) potentially impacted by an intergenic CNV gain (e.g., homozygous duplication).



FIG. 10 represents an example of a gene (VDAC1) potentially impacted by an intergenic CNV (homozygous loss).



FIG. 11 represents an example of genes (EGR1 and ETF1) potentially impacted by an intergenic CNV (homozygous loss).



FIG. 12 represents an example of a gene (ITSN2) potentially impacted by an intergenic CNV (homozygous loss).



FIG. 13 represents an example of known and/or predicted protein interactions using the String database for 21 of 43 genes (non-redundant list) reported in Table 7. The number of PML cases found to harbor variants impacting a given gene is indicated next to each gene.





SUMMARY OF THE INVENTION

Provided herein is a method of treating a condition in a subject in need thereof, comprising: administering a therapeutically effective amount of one or more immunosuppressive medications to the subject, wherein the subject is identified as not having a risk of developing progressive multifocal leukoencephalopathy (PML) by a genetic test. In some embodiments, the subject is identified as not having a high risk of developing PML by a genetic test.


In some embodiments, the condition is a cancer, an organ transplant, or an autoimmune disease.


In some embodiments, the condition is an autoimmune disease.


In some embodiments, the autoimmune disease is selected from the group consisting of Addison disease, Anti-NMDA receptor encephalitis, antisynthetase syndrome, Aplastic anemia, autoimmune anemias, Autoimmune hemolytic anemia, Autoimmune pancreatitis, Behcet's Disease, bullous skin disorders, Celiac disease—sprue (gluten-sensitive enteropathy), chronic fatigue syndrome, Chronic inflammatory demyelinating polyneuropathy, chronic lymphocytic leukemia, Crohn's disease, Dermatomyositis, Devic's disease, Erythroblastopenia, Evans syndrome, Focal segmental glomerulosclerosis, Granulomatosis with polyangiitis, Graves disease, Graves' ophthalmopathy, Guillain-Barre syndrome, Hashimoto thyroiditis, idiopathic thrombocytopenic purpura (ITP), IgA nephropathy, IgA-mediated autoimmune diseases, IgG4-related disease, Inflammatory bowel disease, Juvenile idiopathic arthritis, Multiple sclerosis, Myasthenia gravis, myeloma, non-Hodgkin's lymphoma, Opsoclonus myoclonus syndrome (OMS), Pemphigoid, Pemphigus, pemphigus vulgaris, Pernicious anemia, polymyositis, Psoriasis, pure red cell aplasia, Reactive arthritis, Rheumatoid arthritis, Sarcoidosis, scleroderma, Sjögren syndrome, Systemic lupus erythematosus, Thrombocytopenic purpura, Thrombotic thrombocytopenic purpura, Type I diabetes, Ulcerative colitis, Vasculitis (e.g., vasculitis associated with anti-neutrophil cytoplasmic antibody), Vitiligo, and combinations thereof.


In some embodiments, the autoimmune disease is multiple sclerosis or Crohn's disease. In some embodiments, the autoimmune disease is multiple sclerosis. In some embodiments, the multiple sclerosis is a relapsing form of multiple sclerosis. In some embodiments, the multiple sclerosis is relapsing-remitting multiple sclerosis (RRMS). In some embodiments, the multiple sclerosis is primary progressive multiple sclerosis (PPMS). In some embodiments, the multiple sclerosis is secondary progressive multiple sclerosis (SPMS).


In some embodiments, the one or more immunosuppressive medications comprise a glucocorticoid, cytostatic, antibody, drug acting on immunophilins, interferon, opioid, TNF binding protein, mycophenolate, small biological agent, small molecule, organic compound, or any combination thereof.


In some embodiments, the one or more immunosuppressive medications comprise abatacept, adalimumab, alefacept, alemtuzumab, anakinra, azathioprine, belimumab, bendamustine, bevacizumab, bortezomib (e.g., Velcade), eculizumab (e.g., Soliris), leflunomide, brentuximab vedotin, capecitabine, carboplatin, cetuximab, chlorambucil, cladribine, cyclophosphamide, cyclosporine, daclizumab, doxorubicin, efalizumab, etanercept, etoposide, fludarabine, gemcitabine, ibritumomab tiuxetan, imatinib, infliximab, lenalidomide, methotrexate, mycophenolate mofetil, natalizumab, oxaliplatin, rituximab, tocilizumab, tofacitinib, ustekinumab, vedolizumab, vincristine, belatacept, cytotoxic chemotherapy, corticosteroids, antithymocyte Ig, basiliximab, muromonab-CD3, mycophenolic acid, prednisone/prednisolone, sirolimus (rapamycin), tacrolimus, dimethyl fumarate, fingolimod, ruxolitinib, interferon beta-1a, interferon beta-1b, glatiramer acetate, peginterferon beta-1a, teriflunomide, mitoxantrone, ocrelizumab, asparaginase, bleomycin, busulfan, carmustine, certolizumab, ibrutinib, idarubicin, idelalisib, hydrocortisone, ifosfamide, levamisole, mercaptopurine, mizoribine, obinutuzumab, ofatumumab, tegafur/gimeracil/oteracil, thiotepa, vinblastine, or any combination thereof.


In some embodiments, the one or more immunosuppressive medications comprise interferon beta-1a, interferon beta-1b, glatiramer acetate, peginterferon beta-1a, teriflunomide, fingolimod, dimethyl fumarate, alemtuzumab, mitoxantrone, natalizumab, daclizumab, ocrelizumab, or any combination thereof.


In some embodiments, the subject has not taken the one or more immunosuppressive medications. In some embodiments, the subject has taken the one or more immunosuppressive medications. In some embodiments, the subject is taking the one or more immunosuppressive medications.


In some embodiments, the one or more immunosuppressive medications comprise natalizumab (Tysabri). In some embodiments, at least about 10 mg of the natalizumab is administered, for example, at least about 10 mg, at least about 15 mg, at least about 20 mg, at least about 30 mg, at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 150 mg, at least about 200 mg, at least about 250 mg, or at least about 300 mg of the natalizumab is administered. In some embodiments, at least about 10 mg of the natalizumab is administered via intravenous infusion. In some embodiments, at least about 10 mg of the natalizumab is administered via intravenous infusion in four weeks.


In some embodiments, about 100 mg to about 500 mg of the natalizumab is administered, for example, about 100 mg to about 200 mg, about 100 mg to about 300 mg, about 100 mg to about 400 mg, about 100 mg to about 500 mg, about 200 mg to about 300 mg, about 200 mg to about 400 mg, about 200 mg to about 500 mg, about 300 mg to about 400 mg, about 300 mg to about 500 mg, or about 400 mg to about 500 mg of the natalizumab is administered. In some embodiments, about 100 mg to about 500 mg of the natalizumab is administered via intravenous infusion. In some embodiments, about 100 mg to about 500 mg of the natalizumab is administered via intravenous infusion in four weeks. In some embodiments, about 300 mg of the natalizumab is administered. In some embodiments, about 300 mg of the natalizumab is administered via intravenous infusion. In some embodiments, about 300 mg of the natalizumab is administered via intravenous infusion in four weeks.


In some embodiments, the subject does not have one or more genetic variations associated with a risk of developing PML. In some embodiments, the subject does not have one or more genetic variations associated with a high risk of developing PML.


In some embodiments, the genetic test comprises detecting one or more genetic variations associated with a risk of developing PML in a polynucleic acid sample from the subject. In some embodiments, the genetic test comprises detecting one or more genetic variations associated with a high risk of developing PML in a polynucleic acid sample from the subject.


In some embodiments, the one or more genetic variations comprise a point mutation, polymorphism, single nucleotide polymorphism (SNP), single nucleotide variation (SNV), translocation, insertion, deletion, amplification, inversion, interstitial deletion, copy number variation (CNV), loss of heterozygosity, or any combination thereof.


In some embodiments, the one or more genetic variations disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a method of treating a condition in a subject in need of natalizumab therapy, comprising: administering a therapeutically effective amount of natalizumab to the subject, wherein the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a method of reducing a risk of a subject developing progressive multifocal leukoencephalopathy (PML) comprising administering a therapeutically effective amount of natalizumab to the subject, wherein the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


In some embodiments, the condition is multiple sclerosis.


In some embodiments, the condition is Crohn's disease.


Provided herein is a method of treating multiple sclerosis comprising administering natalizumab to a subject with multiple sclerosis, wherein the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a method of treating Crohn's disease comprising administering natalizumab to a subject with Crohn's disease, wherein the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a method of treating multiple sclerosis comprising testing a subject with multiple sclerosis for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject does not have the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and


administering natalizumab to the subject that was determined not to have the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a method of treating Crohn's disease comprising testing a subject with Crohn's disease for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject does not have the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and administering natalizumab to the subject that was determined not to have the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a method of reducing a risk of a subject developing progressive multifocal leukoencephalopathy (PML) comprising testing a subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject has at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and advising against administering natalizumab to the subject that was determined to have at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


In some embodiments, the subject has multiple sclerosis.


In some embodiments, the subject has Crohn's disease.


Provided herein is a method of treating multiple sclerosis comprising testing a subject with multiple sclerosis for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject has at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and advising against administering natalizumab to the subject that was determined to have at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a method of treating Crohn's disease comprising testing a subject with Crohn's disease for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject has at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and advising against administering natalizumab to the subject that was determined to have at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


In some embodiments, the advising comprises advising that administering natalizumab is contraindicated.


In some embodiments, the advising comprises advising that administering natalizumab increases the risk of the subject developing progressive multifocal leukoencephalopathy (PML)


In some embodiments, the advising comprises advising that administering natalizumab is a factor that increases the risk of the subject developing progressive multifocal leukoencephalopathy (PML).


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Table 13.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Table 14.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Table 15.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Table 16.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Table 17.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Table 18.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene selected from the group consisting of ALG12, AP3B1, ASH1L, ATL2, ATM, ATR, BACH1, BLM, CHD7, CLCN7, CR2, CX3CR1, DOCK2, DOCK8, EHF, EPG5, FAS, FUK, GFI1, GOLGB1, GTPBP4, HIVEP1, HIVEP2, HIVEP3, IFIH1, IGLL1, IL10, IL12B, IL17F, ITK, ITSN2, JAGN1, KITLG, LRBA, LYST, MALT1, MAVS, MCEE, NHEJ1, NOD2, NRIP1, ORAI1, PGM3, PIK3CD, PLCG2, PNP, POLE, PRF1, RBCK1, RBFOX1, RNASEL, RTEL1, SALL2, SHARPIN, SNAP29, STIM2, STXBP2, TAP1, TBC1D16, TCIRG1, TICAM1, TMEM173, TNFRSF10A, TTC7A, VPS13B, and combinations thereof.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene selected from the group consisting of PLCG2, RBCK1, EPG5, IL17F, SHARPIN, PRF1, JAGN1, TAP1, POLE, LRBA, EHF, IL12B, ATL2, NHEJ1, LYST, HIVEP1, AP3B1, TNFRSF10A, PIK3CD, PNP, MCEE, DOCK2, ALG12, and combinations thereof.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene selected from the group consisting of PLCG2, IFIH1, TCIRG1, IGLL1, MAVS, SHARPIN, CHD7, CX3CR1, LRBA, HIVEP3, RNASEL, and combinations thereof.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene selected from the group consisting of SHARPIN, RTEL1, PGM3, TMEM173, CLCN7, MAVS, ORAI1, RBFOX1, MALT1, GFI1, DOCK2, ATM, SNAP29, TICAM1, GTPBP4, BACH1, STXBP2, FAS, GOLGB1, FUK, IL10, ITK, STIM2, ASH1L, TBC1D16, LYST, SALL2, CHD7, BLM, NOD2, IGLL1, TTC7A, KITLG, ATR, ATM, CR2, HIVEP2, ITSN2, DOCKS, VPS13B, NRIP1, and combinations thereof.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene selected from the group consisting of SHARPIN, IFIH1, PLCG2, CHD7, and combinations thereof.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene selected from the group consisting of PLCG2, POLE, LRBA, EPG5, SHARPIN, and combinations thereof.


In some embodiments, the testing comprises testing the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene selected from the group consisting of PLCG2, CHD7, IFIH1, AP3B1, EPG5, PIK3CD, LRBA, SHARPIN, and combinations thereof.


In some embodiments, the subject is identified as not having a risk of developing progressive multifocal leukoencephalopathy (PML) by a genetic test. In some embodiments, the subject is identified as not having a high risk of developing progressive multifocal leukoencephalopathy (PML) by a genetic test.


In some embodiments, the testing comprises assaying a polynucleic acid sample from the subject for the one or more genetic variations.


In some embodiments, the one or more genetic variations result in a loss of function of the corresponding gene.


In some embodiments, the corresponding gene comprises a gene selected from the group consisting of gene numbers (GNs) GN1-GN490.


In some embodiments, the corresponding gene comprises a gene selected from the group consisting of gene numbers (GNs) 1-156 (in Table 3).


In some embodiments, the corresponding gene comprises a gene selected from the group consisting of gene numbers (GNs) in Table 6.


In some embodiments, the corresponding gene comprises a gene selected from the group consisting of PLCG2, RBCK1, EPG5, IL17F, SHARPIN, PRF1, JAGN1, TAP1, POLE, LRBA, EHF, IL12B, ATL2, NHEJ1, LYST, HIVEP1, AP3B1, TNFRSF10A, PIK3CD, PNP, MCEE, DOCK2 and ALG12 (see Table 13).


In some embodiments, the one or more genetic variations are encoded by a sequence with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NOs 1-172 or SRN1-SRN363, with 100% sequence identity to SEQ ID NOs 1000-1329, or with at least 80% and less than 100% sequence identity to GN1-GN490, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NOs 1-172, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV sub-region (SRN) with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SRN1-SRN363, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NOs: 1000-1329, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a sequence with at least 80% and less than 100% sequence identity to GN1-GN490, or complements thereof


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1000, 1001, 1002, 1009, 1010, 1011, 1012, 1014, 1016, 1017, 1019, 1020, 1028, 1032, 1033, 1034, 1035, 1036, 1037, 1040, 1041, 1043, 1051, 1054, 1056, 1057, 1058, 1059, 1061, 1062, 1063, 1066, 1068, 1069, 1070, 1071, 1073, 1074, 1075, 1076, 1077, 1078, 1080, 1082, 1084, 1090, 1092, 1098, 1099, 1100, 1101, 1104, 1107, 1114, 1116, 1118, 1121, 1122, 1123, 1125, 1126, 1127, 1128, 1129, 1130, 1131, 1133, 1135, 1136, 1137, 1138, 1142, 1146, 1147, 1148, 1150, 1152, 1154, 1157, 1160, 1161, 1165, 1166, 1167, 1168, 1169, 1171, 1174, 1175, 1176, 1177, 1178, 1179, 1180, 1181, 1182, 1183, 1184, 1193, 1194, 1200, 1201, 1202, 1203, 1204, 1208, 1219, 1220, 1221, 1222, 1226, 1227, 1228, 1229, 1230, 1231, 1232, 1235, 1239, 1247, 1248, 1249, 1250, 1251, 1252, 1254, 1255, 1256, 1259, 1260, 1261, 1263, 1264, 1266, 1267, 1273, 1278, 1279, 1283, 1284, 1286, 1287, 1289, 1290, 1291, 1299, 1300, 1301, 1304, 1311, 1327 or 1328 (see Tables 7 and 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1011, 1020, 1028, 1032, 1034, 1035, 1036, 1040, 1056, 1069, 1073, 1077, 1101, 1114, 1123, 1125, 1126, 1127, 1135, 1142, 1146, 1147, 1148, 1152, 1154, 1157, 1167, 1174, 1184, 1193, 1194, 1203, 1208, 1221, 1222, 1229, 1235, 1252, 1255, 1256, 1259, 1260, 1261, 1263, 1273, 1278, 1279, 1284, 1287, 1289, 1299 or 1311 (see Table 7), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1000, 1001, 1002, 1009, 1010, 1012, 1014, 1016, 1017, 1019, 1033, 1037, 1041, 1043, 1051, 1054, 1057, 1058, 1059, 1061, 1062, 1063, 1066, 1068, 1070, 1071, 1074, 1075, 1076, 1078, 1080, 1082, 1084, 1090, 1092, 1098, 1099, 1100, 1104, 1107, 1116, 1118, 1121, 1122, 1128, 1129, 1130, 1131, 1133, 1136, 1137, 1138, 1146, 1147, 1150, 1152, 1160, 1161, 1165, 1166, 1168, 1169, 1171, 1175, 1176, 1177, 1178, 1179, 1180, 1181, 1182, 1183, 1200, 1201, 1202, 1204, 1219, 1220, 1226, 1227, 1228, 1230, 1231, 1232, 1239, 1247, 1248, 1249, 1250, 1251, 1252, 1254, 1264, 1266, 1267, 1278, 1279, 1283, 1286, 1290, 1291, 1300, 1301, 1304, 1327 or 1328 (see Table 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr16:81942175 A>G, chr2:163136505 C>G, chr11:67818269 G>A, chr22:23917192 G>T, chr20:3846397 C>T, chr8:145154222, G>A chr8:61654298 T>A, chr3:39323163 A>C, chr4:151199080 G>A, chr1:42047208 C>G, chr2:163124051 C>T, chr1:182554557 C>T, chr8:145154824 A>C, chr20:62305450 C>T, chr22:23915745 G>A, chr6:83884161 C>G, chr11:108202772 G>T, chr5:138856923 C>T, chr16:1510535 C>T, chr20:3843027 C>A, chr12:122064788 G>GT, chr16:7714909 C>T, chr18:56401523 C>T, chr1:92946625 G>C, chr5:169081453 G>C, chr11:108117787 C>T, chr22:21235389 A>G, chr19:4817657 C>T, chr10:1060218 G>A, chr21:30698953 T>G, chr9:304628 G>A, chr19:7712287 G>C, chr10:90771767 G>A, chr3:121415370 T>C, chr16:70503095 A>G, chr1:206945738 C>T, chr5:156593120 C>T, chr4:27019452 C>T, chr1:155317682 C>T, chr17:77926526 C>T, chr1:235840495 G>T, chr14:21993359 G>A, chr8:61757805 C>T, chr15:91306241 G>A, chr16:50741791 C>T, chr22:23915583 T>C, chr2:47205921 C>T, chr12:88900891 C>A, chr3:142281353 C>G, chr11:108123551 C>T, chr1: 207641950 C>T, chr6:143092151 T>C, chr2:24431184 C>T, chr2:24432937 C>T, chr9:312134 G>A, chr8:100205255 G>A, chr21:16339852 T>C, and any combination thereof (see Tables 14 and 15).


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr16:81942175 A>G, chr2:163136505 C>G, chr11:67818269 G>A, chr22:23917192 G>T, chr20:3846397 C>T, chr8:145154222, G>A chr8:61654298 T>A, chr3:39323163 A>C, chr4:151199080 G>A, chr1:42047208 C>G, chr2:163124051 C>T, chr1:182554557 C>T, and any combination thereof (see Table 14).


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr8:145154824 A>C, chr20:62305450 C>T, chr22:23915745 G>A, chr6:83884161 C>G, chr11:108202772 G>T, chr5:138856923 C>T, chr16:1510535 C>T, chr20:3843027 C>A, chr12:122064788 G>GT, chr16:7714909 C>T, chr18:56401523 C>T, chr1:92946625 G>C, chr5:169081453 G>C, chr11:108117787 C>T, chr22:21235389 A>G, chr19:4817657 C>T, chr10:1060218 G>A, chr21:30698953 T>G, chr9:304628 G>A, chr19:7712287 G>C, chr10:90771767 G>A, chr3:121415370 T>C, chr16:70503095 A>G, chr1:206945738 C>T, chr5:156593120 C>T, chr4:27019452 C>T, chr1:155317682 C>T, chr17:77926526 C>T, chr1:235840495 G>T, chr14:21993359 G>A, chr8:61757805 C>T, chr15:91306241 G>A, chr16:50741791 C>T, chr22:23915583 T>C, chr2:47205921 C>T, chr12:88900891 C>A, chr3:142281353 C>G, chr11:108123551 C>T, chr1:207641950 C>T, chr6:143092151 T>C, chr2:24431184 C>T, chr2:24432937 C>T, chr9:312134 G>A, chr8:100205255 G>A, chr21:16339852 T>C, and any combination thereof (see Table 15).


In some embodiments, the SNV is a heterozygous SNV.


In some embodiments, the SNV is a homozygous SNV.


In some embodiments, the one or more genetic variations comprise a pair of single nucleotide variations (SNVs), wherein the pair of SNVs are encoded by any one of SEQ ID NO pairs: 1003 and 1004, 1003 and 1005, 1006 and 1007, 1024 and 1025, 1030 and 1031, 1047 and 1048, 1049 and 1050, 1063 and 1064, 1063 and 1065, 1063 and 1066, 1075 and 1076, 1091 and 1093, 1091 and 1096, 1093 and 1095, 1094 and 1097, 1098 and 1099, 1098 and 1100, 1099 and 1100, 1102 and 1103, 1104 and 1106, 1104 and 1107, 1104 and 1108, 1104 and 1109, 1104 and 1110, 1104 and 1111, 1104 and 1112, 1110 and 1111, 1112 and 1113, 1119 and 1120, 1124 and 1125, 1124 and 1126, 1125 and 1126, 1140 and 1141, 1142 and 1144, 1146 and 1151, 1147 and 1148, 1147 and 1149, 1153 and 1146, 1153 and 1147, 1155 and 1156, 1160 and 1161, 1165 and 1166, 1186 and 1187, 1188 and 1193, 1189 and 1193, 1191 and 1192, 1191 and 1193, 1191 and 1195, 1192 and 1193, 1192 and 1195, 1196 and 1197, 1206 and 1207, 1210 and 1218, 1211 and 1213, 1212 and 1213, 1213 and 1215, 1213 and 1216, 1213 and 1217, 1233 and 1238, 1242 and 1243, 1245 and 1246, 1263 and 1260, 1269 and 1279, 1270 and 1279, 1270 and 1282, 1271 and 1279, 1274 and 1279, 1278 and 1279, 1278 and 1281, 1279 and 1280, 1279 and 1281, 1279 and 1282, 1292 and 1293, 1296 and 1297, 1305 and 1314, 1306 and 1310, 1313 and 1321 or 1315 and 1322 (see Table 9 or Tables 9 and 7 for a subset), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 157, 2, 140, 65, 26, 14 or 45 (see Tables 7 and 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 2, 140, 65, 26, 14 or 45 (see Table 7), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NO 157 (see Table 8), or a complement thereof


In some embodiments, the one or more genetic variations comprise a CNV-SNV pair comprising a CNV and a single nucleotide variation (SNV), wherein the SNV of the CNV-SNV pair is encoded by any one of SEQ ID NO pairs: 146 and 1301, 85 and 1173, 58 and 1107, 58 and 1104, 91 and 1199, 103 and 1225, 103 and 1086 or 41 and 1223 (see Tables 1 and 10), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of: chr8:145154222 G>A, chr2:163136505 C>G, chr16:81942175 A>G, chr8:61654298 T>A, and combinations thereof (see Tables 14 and 16).


In some embodiments, the one or more genetic variations disrupt or modulate one or more of the following genes: PLCG2, POLE, LRBA, EPG5 and SHARPIN (see Table 17).


In some embodiments, the one or more genetic variations disrupt or modulate one or more of the following genes: PLCG2, CHD7, IFIH1, AP3B1, EPG5, PIK3CD, LRBA and SHARPIN (see Table 18).


In some embodiments, the corresponding gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 173-455 or 1500-2177 (see Tables 4 and 12), or complements thereof.


In some embodiments, the corresponding gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 173-455 (see Table 4), or complements thereof.


In some embodiments, the corresponding gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 1500-2177 (see Table 12), or complements thereof.


In some embodiments, the one or more genetic variations comprise 2 or 3 or 4 or 5 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 10 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 20 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 50 or more genetic variations.


In some embodiments, the genetic test or the testing comprises microarray analysis, PCR, sequencing, nucleic acid hybridization, or any combination thereof.


In some embodiments, the genetic test or the testing comprises microarray analysis selected from the group consisting of a Comparative Genomic Hybridization (CGH) array analysis and an SNP array analysis.


In some embodiments, the genetic test or the testing comprises sequencing, wherein the sequencing is selected from the group consisting of Massively Parallel Signature Sequencing (MPSS), polony sequencing, 454 pyrosequencing, Illumina sequencing, Illumina (Solexa) sequencing using 10× Genomics library preparation, SOLiD sequencing, ion semiconductor sequencing, DNA nanoball sequencing, heliscope single molecule sequencing, single molecule real time (SMRT) sequencing, RNAP sequencing, Nanopore DNA sequencing, sequencing by hybridization, and microfluidic Sanger sequencing.


In some embodiments, the genetic test or the testing comprises analyzing a whole genome of the subject.


In some embodiments, the genetic test or the testing comprises analyzing a whole exome of the subject.


In some embodiments, the genetic test or the testing comprises analyzing nucleic acid information that has already been obtained for a whole genome or a whole exome of the subject.


In some embodiments, the nucleic acid information is obtained from an in silico analysis.


In some embodiments, the subject is a human subject.


In some embodiments, the polynucleic acid sample comprises a polynucleic acid from blood, saliva, urine, serum, tears, skin, tissue, or hair of the subject.


In some embodiments, the method further comprises treating the subject with an agent that reduces a viral load in the subject.


In some embodiments, the immunosuppressive agent is administered after the viral load is reduced.


In some embodiments, the viral load is a JCV viral load.


In some embodiments, the agent that reduces the viral load is an agent that targets JCV.


In some embodiments, the method further comprises analyzing for a presence of JCV in a biological sample from the subject. In some embodiments, the method comprises a JCV-antibody test. In some embodiments, the JCV-antibody test has a negative result. In some embodiments, the JCV-antibody test does not detect a presence of JCV in the biological sample from the subject. In some embodiments, the JCV-antibody test detects a presence of JCV in the biological sample from the subject.


In some embodiments, the analyzing for a presence of JCV comprises contacting a JCV detection reagent to the biological sample.


In some embodiments, the JCV detection reagent is selected from the group consisting of an anti-JCV antibody, a JCV specific primer, and combinations thereof.


Provided herein is a method of treating a condition in a subject in need thereof, comprising: administering a therapeutically effective amount of one or more immunosuppressive medications to the subject, and one or more agents that reduce a viral load in the subject, wherein the subject is identified as not having a risk of developing progressive multifocal leukoencephalopathy (PML) by a genetic test. In some embodiments, the subject is identified as not having a high risk of developing progressive multifocal leukoencephalopathy (PML) by a genetic test.


Provided herein is a method of treating a condition in a subject in need thereof, comprising: analyzing a polynucleic acid sample from the subject for one or more genetic variations that disrupt or modulate a gene of GN1-GN490, wherein a genetic variation of the one or more genetic variations that disrupt or modulate a gene of GN1-GN490 is not present in the polynucleic acid sample; identifying the subject as not having a risk of developing PML; administering a therapeutically effective amount of one or more immunosuppressive medications to the subject. In some embodiments, the method comprises identifying the subject as not having a high risk of developing PML.


Provided herein is a method of identifying a subject as having a risk of developing PML, comprising: analyzing a polynucleic acid sample from the subject for one or more genetic variations that disrupt or modulate a gene of GN1-GN490, wherein a genetic variation of the one or more genetic variations that disrupt or modulate a gene of GN1-GN490 is not present in the polynucleic acid sample; identifying the subject as not having a risk of developing PML. In some embodiments, the method comprises identifying the subject as not having a high risk of developing PML.


Provided herein is a method of identifying a subject as having a risk of developing progressive multifocal leukoencephalopathy (PML) comprising obtaining a genetic test result from a polynucleic acid sample from a subject, and identifying the subject as having a risk of developing PML based on the genetic test result; wherein the subject is immunosuppressed.


Provided herein is a method of monitoring a subject as having a risk of developing progressive multifocal leukoencephalopathy (PML) comprising obtaining a genetic test result from a polynucleic acid sample from a subject, and identifying the subject as having an increased risk of developing PML based on the genetic test result; wherein the subject is immunosuppressed.


In some embodiments, the subject is on an immunosuppressive therapy.


Provided herein is a method of identifying a subject as having a risk of developing progressive multifocal leukoencephalopathy (PML) comprising detecting one or more genetic variations that disrupt or modulate a gene of GN1-GN490 in a polynucleic acid sample from a subject, and identifying the subject as having a risk of developing PML; wherein the subject is immunosuppressed.


Provided herein is a method of identifying a subject as having a risk of developing progressive multifocal leukoencephalopathy (PML) comprising: analyzing a polynucleic acid sample from the subject for one or more genetic variations that disrupt or modulate a gene of GN1-GN490, wherein a genetic variation of the one or more genetic variations that disrupt or modulate a gene of GN1-GN490 is present in the polynucleic acid sample; identifying the subject as having a risk of developing PML; wherein the subject is immunosuppressed. In some embodiments, the method comprises identifying the subject as having a high risk of developing PML.


In some embodiments, the subject has HIV. In some embodiments, the subject has HIV infection. In some embodiments, the subject is at risk of HIV infection.


In some embodiments, the condition is a cancer, a hematologic malignancy, an organ transplant, or an autoimmune disease. In some embodiments, the condition is idiopathic CD4+ lymphocytopenia (ICL).


In some embodiments, the condition is an autoimmune disease.


In some embodiments, the autoimmune disease is selected from the group consisting of Addison disease, Behcet's Disease, Inflammatory bowel disease, Celiac disease-sprue (gluten-sensitive enteropathy), Crohn's disease, Dermatomyositis, Focal segmental glomerulosclerosis, Graves disease, Hashimoto thyroiditis, Multiple sclerosis, Myasthenia gravis, Pemphigus, Pemphigoid, Aplastic anemia, Pernicious anemia, Autoimmune hemolytic anemia, Erythroblastopenia, Thrombocytopenic purpura, Evans syndrome, Vasculitis, Granulomatosis with polyangiitis, Chronic inflammatory demyelinating polyneuropathy, Guillain-Barre syndrome, Anti-NMDA receptor encephalitis, Devic's disease, Autoimmune pancreatitis, Opsoclonus myoclonus syndrome, IgG4-related disease, Psoriasis, Reactive arthritis, Rheumatoid arthritis, Juvenile idiopathic arthritis, Sarcoidosis, Sjögren syndrome, Systemic lupus erythematosus, Type I diabetes, Vitiligo, or Ulcerative colitis.


In some embodiments, the autoimmune disease is multiple sclerosis or Crohn's disease.


In some embodiments, the one or more immunosuppressive medications comprise a glucocorticoid, cytostatic, antibody, drug acting on immunophilins, interferon, opioid, TNF binding protein, mycophenolate, small biological agent, small molecule, organic compound, or any combination thereof.


In some embodiments, the one or more immunosuppressive medications comprise a interferon beta-1a, interferon beta-1b, glatiramer acetate, peginterferon beta-1a, teriflunomide, fingolimod, dimethyl fumarate, alemtuzumab, mitoxantrone, natalizumab, daclizumab, ocrelizumab, or any combination thereof.


In some embodiments, the one or more immunosuppressive medications comprise natalizumab (Tysabri).


In some embodiments, the one or more genetic variations comprise a point mutation, polymorphism, single nucleotide polymorphisms (SNP), single nucleotide variation (SNV), translocation, insertion, deletion, amplification, inversion, interstitial deletion, copy number variation (CNV), loss of heterozygosity, or any combination thereof.


In some embodiments, the one or more genetic variations result in a loss of function of the corresponding gene.


In some embodiments, the corresponding gene comprises a gene selected from the group consisting of gene numbers (GNs) GN1-GN490.


In some embodiments, the gene comprises a gene selected from the group consisting of gene numbers (GNs) 1-156 (in Table 3).


In some embodiments, the gene comprises a gene selected from the group consisting of gene numbers (GNs) in Table 6.


In some embodiments, the gene comprises a gene selected from the group consisting of PLCG2, RBCK1, EPG5, IL17F, SHARPIN, PRF1, JAGN1, TAP1, POLE, LRBA, EHF, IL12B, ATL2, NHEJ1, LYST, HIVEP1, AP3B1, TNFRSF10A, PIK3CD, PNP, MCEE, DOCK2 and ALG12 (see Table 13).


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a sequence with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NOs 1-172 or SRN1-SRN363, with 100% sequence identity to SEQ ID NOs 1000-1329, or with at least 80% and less than 100% sequence identity to GN1-GN490, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NOs 1-172, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV sub-region (SRN) with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SRN1-SRN363, or complements thereof.


In some embodiments, the one or more genetic variations are encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NOs: 1000-1329, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a sequence with at least 80% and less than 100% sequence identity to GN1-GN490, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1000, 1001, 1002, 1009, 1010, 1011, 1012, 1014, 1016, 1017, 1019, 1020, 1028, 1032, 1033, 1034, 1035, 1036, 1037, 1040, 1041, 1043, 1051, 1054, 1056, 1057, 1058, 1059, 1061, 1062, 1063, 1066, 1068, 1069, 1070, 1071, 1073, 1074, 1075, 1076, 1077, 1078, 1080, 1082, 1084, 1090, 1092, 1098, 1099, 1100, 1101, 1104, 1107, 1114, 1116, 1118, 1121, 1122, 1123, 1125, 1126, 1127, 1128, 1129, 1130, 1131, 1133, 1135, 1136, 1137, 1138, 1142, 1146, 1147, 1148, 1150, 1152, 1154, 1157, 1160, 1161, 1165, 1166, 1167, 1168, 1169, 1171, 1174, 1175, 1176, 1177, 1178, 1179, 1180, 1181, 1182, 1183, 1184, 1193, 1194, 1200, 1201, 1202, 1203, 1204, 1208, 1219, 1220, 1221, 1222, 1226, 1227, 1228, 1229, 1230, 1231, 1232, 1235, 1239, 1247, 1248, 1249, 1250, 1251, 1252, 1254, 1255, 1256, 1259, 1260, 1261, 1263, 1264, 1266, 1267, 1273, 1278, 1279, 1283, 1284, 1286, 1287, 1289, 1290, 1291, 1299, 1300, 1301, 1304, 1311, 1327 or 1328 (see Tables 7 and 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1011, 1020, 1028, 1032, 1034, 1035, 1036, 1040, 1056, 1069, 1073, 1077, 1101, 1114, 1123, 1125, 1126, 1127, 1135, 1142, 1146, 1147, 1148, 1152, 1154, 1157, 1167, 1174, 1184, 1193, 1194, 1203, 1208, 1221, 1222, 1229, 1235, 1252, 1255, 1256, 1259, 1260, 1261, 1263, 1273, 1278, 1279, 1284, 1287, 1289, 1299 or 1311 (see Table 7), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1000, 1001, 1002, 1009, 1010, 1012, 1014, 1016, 1017, 1019, 1033, 1037, 1041, 1043, 1051, 1054, 1057, 1058, 1059, 1061, 1062, 1063, 1066, 1068, 1070, 1071, 1074, 1075, 1076, 1078, 1080, 1082, 1084, 1090, 1092, 1098, 1099, 1100, 1104, 1107, 1116, 1118, 1121, 1122, 1128, 1129, 1130, 1131, 1133, 1136, 1137, 1138, 1146, 1147, 1150, 1152, 1160, 1161, 1165, 1166, 1168, 1169, 1171, 1175, 1176, 1177, 1178, 1179, 1180, 1181, 1182, 1183, 1200, 1201, 1202, 1204, 1219, 1220, 1226, 1227, 1228, 1230, 1231, 1232, 1239, 1247, 1248, 1249, 1250, 1251, 1252, 1254, 1264, 1266, 1267, 1278, 1279, 1283, 1286, 1290, 1291, 1300, 1301, 1304, 1327 or 1328 (see Table 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr16:81942175 A>G, chr2:163136505 C>G, chr11:67818269 G>A, chr22:23917192 G>T, chr20:3846397 C>T, chr8:145154222, G>A chr8:61654298 T>A, chr3:39323163 A>C, chr4:151199080 G>A, chr1:42047208 C>G, chr2:163124051 C>T, chr1:182554557 C>T, chr8:145154824 A>C, chr20:62305450 C>T, chr22:23915745 G>A, chr6:83884161 C>G, chr11:108202772 G>T, chr5:138856923 C>T, chr16:1510535 C>T, chr20:3843027 C>A, chr12:122064788 G>GT, chr16:7714909 C>T, chr18:56401523 C>T, chr1:92946625 G>C, chr5:169081453 G>C, chr11:108117787 C>T, chr22:21235389 A>G, chr19:4817657 C>T, chr10:1060218 G>A, chr21:30698953 T>G, chr9:304628 G>A, chr19:7712287 G>C, chr10:90771767 G>A, chr3:121415370 T>C, chr16:70503095 A>G, chr1:206945738 C>T, chr5:156593120 C>T, chr4:27019452 C>T, chr1:155317682 C>T, chr17:77926526 C>T, chr1:235840495 G>T, chr14:21993359 G>A, chr8:61757805 C>T, chr15:91306241 G>A, chr16:50741791 C>T, chr22:23915583 T>C, chr2:47205921 C>T, chr12:88900891 C>A, chr3:142281353 C>G, chr11:108123551 C>T, chr1:207641950 C>T, chr6:143092151 T>C, chr2:24431184 C>T, chr2:24432937 C>T, chr9:312134 G>A, chr8:100205255 G>A, chr21:16339852 T>C, and any combination thereof (see Tables 14 and 15).


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr16:81942175 A>G, chr2:163136505 C>G, chr11:67818269 G>A, chr22:23917192 G>T, chr20:3846397 C>T, chr8:145154222, G>A chr8:61654298 T>A, chr3:39323163 A>C, chr4:151199080 G>A, chr1:42047208 C>G, chr2:163124051 C>T, chr1:182554557 C>T, and any combination thereof (see Table 14).


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr8:145154824 A>C, chr20:62305450 C>T, chr22:23915745 G>A, chr6:83884161 C>G, chr11:108202772 G>T, chr5:138856923 C>T, chr16:1510535 C>T, chr20:3843027 C>A, chr12:122064788 G>GT, chr16:7714909 C>T, chr18:56401523 C>T, chr1:92946625 G>C, chr5:169081453 G>C, chr11:108117787 C>T, chr22:21235389 A>G, chr19:4817657 C>T, chr10:1060218 G>A, chr21:30698953 T>G, chr9:304628 G>A, chr19:7712287 G>C, chr10:90771767 G>A, chr3:121415370 T>C, chr16:70503095 A>G, chr1:206945738 C>T, chr5:156593120 C>T, chr4:27019452 C>T, chr1:155317682 C>T, chr17:77926526 C>T, chr1:235840495 G>T, chr14:21993359 G>A, chr8:61757805 C>T, chr15:91306241 G>A, chr16:50741791 C>T, chr22:23915583 T>C, chr2:47205921 C>T, chr12:88900891 C>A, chr3:142281353 C>G, chr11:108123551 C>T, chr1: 207641950 C>T, chr6:143092151 T>C, chr2:24431184 C>T, chr2:24432937 C>T, chr9:312134 G>A, chr8:100205255 G>A, chr21:16339852 T>C, and any combination thereof (see Table 15).


In some embodiments, the SNV is a heterozygous SNV.


In some embodiments, the SNV is a homozygous SNV.


In some embodiments, the one or more genetic variations comprise a pair of single nucleotide variations (SNVs), wherein the pair of SNVs are encoded by any one of SEQ ID NO pairs: 1003 and 1004, 1003 and 1005, 1006 and 1007, 1024 and 1025, 1030 and 1031, 1047 and 1048, 1049 and 1050, 1063 and 1064, 1063 and 1065, 1063 and 1066, 1075 and 1076, 1091 and 1093, 1091 and 1096, 1093 and 1095, 1094 and 1097, 1098 and 1099, 1098 and 1100, 1099 and 1100, 1102 and 1103, 1104 and 1106, 1104 and 1107, 1104 and 1108, 1104 and 1109, 1104 and 1110, 1104 and 1111, 1104 and 1112, 1110 and 1111, 1112 and 1113, 1119 and 1120, 1124 and 1125, 1124 and 1126, 1125 and 1126, 1140 and 1141, 1142 and 1144, 1146 and 1151, 1147 and 1148, 1147 and 1149, 1153 and 1146, 1153 and 1147, 1155 and 1156, 1160 and 1161, 1165 and 1166, 1186 and 1187, 1188 and 1193, 1189 and 1193, 1191 and 1192, 1191 and 1193, 1191 and 1195, 1192 and 1193, 1192 and 1195, 1196 and 1197, 1206 and 1207, 1210 and 1218, 1211 and 1213, 1212 and 1213, 1213 and 1215, 1213 and 1216, 1213 and 1217, 1233 and 1238, 1242 and 1243, 1245 and 1246, 1263 and 1260, 1269 and 1279, 1270 and 1279, 1270 and 1282, 1271 and 1279, 1274 and 1279, 1278 and 1279, 1278 and 1281, 1279 and 1280, 1279 and 1281, 1279 and 1282, 1292 and 1293, 1296 and 1297, 1305 and 1314, 1306 and 1310, 1313 and 1321 or 1315 and 1322 (see Table 9 or Tables 9 and 7 for a subset), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 157, 2, 140, 65, 26, 14 or 45 (see Tables 7 and 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 2, 140, 65, 26, 14 or 45 (see Table 7), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NO 157 (see Table 8), or a complement thereof.


In some embodiments, the one or more genetic variations comprise a CNV-SNV pair comprising a CNV and a single nucleotide variation (SNV), wherein the SNV of the CNV-SNV pair is encoded by any one of SEQ ID NOs 1301, 1173, 1107, 1104, 1199, 1225, 1086 or 1223 (see Table 10), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of one or more of the following: chr8:145154222 G>A, chr2:163136505 C>G, chr16:81942175 A>G, and chr8:61654298 T>A (see Tables 14 and 16).


In some embodiments, the one or more genetic variations disrupt or modulate one or more of the following genes: PLCG2, POLE, LRBA, EPG5 and SHARPIN (see Table 17).


In some embodiments, the one or more genetic variations disrupt or modulate one or more of the following genes: PLCG2, CHD7, IFIH1, AP3B1, EPG5, PIK3CD, LRBA and SHARPIN (see Table 18).


In some embodiments, the gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 173-455 or 1500-2177 (see Tables 4 and 12), or complements thereof.


In some embodiments, the gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 173-455 (see Table 4), or complements thereof.


In some embodiments, the gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 1500-2177 (see Table 12), or complements thereof.


In some embodiments, the one or more genetic variations comprise 2 or 3 or 4 or 5 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 10 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 20 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 50 or more genetic variations.


In some embodiments, the analyzing comprises microarray analysis, PCR, sequencing, nucleic acid hybridization, or any combination thereof.


In some embodiments, the genetic test result comprises a genetic test result from a microarray analysis, PCR, sequencing, nucleic acid hybridization, or any combination thereof.


In some embodiments, the detecting comprises a microarray analysis, PCR, sequencing, nucleic acid hybridization, or any combination thereof.


In some embodiments, the microarray analysis selected from the group consisting of a Comparative Genomic Hybridization (CGH) array analysis and an SNP array analysis.


In some embodiments, the sequencing is selected from the group consisting of Massively Parallel Signature Sequencing (MPSS), polony sequencing, 454 pyrosequencing, Illumina sequencing, Illumina (Solexa) sequencing using 10× Genomics library preparation, SOLiD sequencing, ion semiconductor sequencing, DNA nanoball sequencing, heliscope single molecule sequencing, single molecule real time (SMRT) sequencing, RNAP sequencing, Nanopore DNA sequencing, sequencing by hybridization, and microfluidic Sanger sequencing.


In some embodiments, the analyzing comprises analyzing a whole genome or a whole exome of the subject.


In some embodiments, the analyzing comprises analyzing nucleic acid information that has already been obtained for a whole genome or a whole exome of the subject.


In some embodiments, the nucleic acid information is obtained from an in silico analysis.


In some embodiments, the analyzing comprises analyzing a whole genome or a whole exome of the subject.


In some embodiments, the analyzing comprises analyzing nucleic acid information that has already been obtained for a whole genome or a whole exome of the subject.


In some embodiments, the nucleic acid information is obtained from an in silico analysis.


In some embodiments, the detecting comprises analyzing a whole genome or a whole exome of the subject.


In some embodiments, the detecting comprises analyzing nucleic acid information that has already been obtained for a whole genome or a whole exome of the subject.


In some embodiments, the nucleic acid information is obtained from an in silico analysis.


In some embodiments, the subject is a human subject.


In some embodiments, the polynucleic acid sample comprises a polynucleic acid from blood, saliva, urine, serum, tears, skin, tissue, or hair of the subject.


In some embodiments, the method further comprises analyzing for a presence of JCV in a biological sample from the subject.


In some embodiments, the analyzing for a presence of JCV comprises contacting a JCV detection reagent to the biological sample.


In some embodiments, the JCV detection reagent is selected from the group consisting of an anti-JCV antibody, a JCV specific primer, and combinations thereof.


Provided herein is a kit, comprising reagents for assaying a polynucleic acid sample from a subject in need thereof for the presence of one or more genetic variations that disrupt or modulate a gene of GN1-GN490.


In some embodiments, the reagents comprise at least one contiguous oligonucleotide that hybridizes to a fragment of the polynucleic acid sample.


In some embodiments, the reagents comprise at least one pair of oligonucleotides that hybridize to opposite strands of a fragment of the polynucleic acid sample.


In some embodiments, the kit further comprises one or more immunosuppressive medications.


In some embodiments, the one or more immunosuppressive medications comprise a glucocorticoid, cytostatic, antibody, drug acting on immunophilins, interferon, opioid, TNF binding protein, mycophenolate, small biological agent, or any combination thereof.


In some embodiments, the one or more immunosuppressive medications comprise a interferon beta-1a, interferon beta-1b, glatiramer acetate, peginterferon beta-1a, teriflunomide, fingolimod, dimethyl fumarate, alemtuzumab, mitoxantrone, natalizumab, daclizumab, ocrelizumab, or any combination thereof.


In some embodiments, the one or more immunosuppressive medications comprise natalizumab (Tysabri).


In some embodiments, the kit further comprises a JCV detection reagent.


In some embodiments, the JCV detection reagent is selected from the group consisting of an anti-JCV antibody, a JCV specific primer, and combinations thereof.


In some embodiments, the kit further comprises a set of instructions for administration of the one or more immunosuppressive medications.


In some embodiments, the one or more genetic variations comprise a point mutation, polymorphism, single nucleotide polymorphisms (SNP), single nucleotide variation (SNV), translocation, insertion, deletion, amplification, inversion, interstitial deletion, copy number variation (CNV), loss of heterozygosity, or any combination thereof.


In some embodiments, the one or more genetic variations result in a loss of function of the corresponding gene.


In some embodiments, the one or more genetic variations comprise 5 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 10 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 20 or more genetic variations.


In some embodiments, the one or more genetic variations comprise 50 or more genetic variations.


In some embodiments, the subject is a human subject.


In some embodiments, the polynucleic acid sample comprises a polynucleic acid from blood, saliva, urine, serum, tears, skin, tissue, or hair of the subject.


Provided herein is a panel of polynucleic acids for detecting one or more genetic variations that disrupt or modulate a gene of GN1-GN490, wherein each polynucleic acid of the panel comprises a sequence complementary to a sequence of one or more genetic variation or complements thereof that disrupts or modulates a gene selected from the group consisting of GN1-GN490.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a sequence with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NOs 1-172 or SRN1-SRN363, with 100% sequence identity to SEQ ID NOs 1000-1329, or with at least 80% and less than 100% sequence identity to GN1-GN490, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NOs 1-172, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV sub-region (SRN) with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SRN1-SRN363, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NOs: 1000-1329, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a sequence with at least 80% and less than 100% sequence identity to GN1-GN490, or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1000, 1001, 1002, 1009, 1010, 1011, 1012, 1014, 1016, 1017, 1019, 1020, 1028, 1032, 1033, 1034, 1035, 1036, 1037, 1040, 1041, 1043, 1051, 1054, 1056, 1057, 1058, 1059, 1061, 1062, 1063, 1066, 1068, 1069, 1070, 1071, 1073, 1074, 1075, 1076, 1077, 1078, 1080, 1082, 1084, 1090, 1092, 1098, 1099, 1100, 1101, 1104, 1107, 1114, 1116, 1118, 1121, 1122, 1123, 1125, 1126, 1127, 1128, 1129, 1130, 1131, 1133, 1135, 1136, 1137, 1138, 1142, 1146, 1147, 1148, 1150, 1152, 1154, 1157, 1160, 1161, 1165, 1166, 1167, 1168, 1169, 1171, 1174, 1175, 1176, 1177, 1178, 1179, 1180, 1181, 1182, 1183, 1184, 1193, 1194, 1200, 1201, 1202, 1203, 1204, 1208, 1219, 1220, 1221, 1222, 1226, 1227, 1228, 1229, 1230, 1231, 1232, 1235, 1239, 1247, 1248, 1249, 1250, 1251, 1252, 1254, 1255, 1256, 1259, 1260, 1261, 1263, 1264, 1266, 1267, 1273, 1278, 1279, 1283, 1284, 1286, 1287, 1289, 1290, 1291, 1299, 1300, 1301, 1304, 1311, 1327 or 1328 (see Tables 7 and 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1011, 1020, 1028, 1032, 1034, 1035, 1036, 1040, 1056, 1069, 1073, 1077, 1101, 1114, 1123, 1125, 1126, 1127, 1135, 1142, 1146, 1147, 1148, 1152, 1154, 1157, 1167, 1174, 1184, 1193, 1194, 1203, 1208, 1221, 1222, 1229, 1235, 1252, 1255, 1256, 1259, 1260, 1261, 1263, 1273, 1278, 1279, 1284, 1287, 1289, 1299 or 1311 (see Table 7), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NO: 1000, 1001, 1002, 1009, 1010, 1012, 1014, 1016, 1017, 1019, 1033, 1037, 1041, 1043, 1051, 1054, 1057, 1058, 1059, 1061, 1062, 1063, 1066, 1068, 1070, 1071, 1074, 1075, 1076, 1078, 1080, 1082, 1084, 1090, 1092, 1098, 1099, 1100, 1104, 1107, 1116, 1118, 1121, 1122, 1128, 1129, 1130, 1131, 1133, 1136, 1137, 1138, 1146, 1147, 1150, 1152, 1160, 1161, 1165, 1166, 1168, 1169, 1171, 1175, 1176, 1177, 1178, 1179, 1180, 1181, 1182, 1183, 1200, 1201, 1202, 1204, 1219, 1220, 1226, 1227, 1228, 1230, 1231, 1232, 1239, 1247, 1248, 1249, 1250, 1251, 1252, 1254, 1264, 1266, 1267, 1278, 1279, 1283, 1286, 1290, 1291, 1300, 1301, 1304, 1327 or 1328 (see Table 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr16:81942175 A>G, chr2:163136505 C>G, chr11:67818269 G>A, chr22:23917192 G>T, chr20:3846397 C>T, chr8:145154222, G>A chr8:61654298 T>A, chr3:39323163 A>C, chr4:151199080 G>A, chr1:42047208 C>G, chr2:163124051 C>T, chr1:182554557 C>T, chr8:145154824 A>C, chr20:62305450 C>T, chr22:23915745 G>A, chr6:83884161 C>G, chr11:108202772 G>T, chr5:138856923 C>T, chr16:1510535 C>T, chr20:3843027 C>A, chr12:122064788 G>GT, chr16:7714909 C>T, chr18:56401523 C>T, chr1:92946625 G>C, chr5:169081453 G>C, chr11:108117787 C>T, chr22:21235389 A>G, chr19:4817657 C>T, chr10:1060218 G>A, chr21:30698953 T>G, chr9:304628 G>A, chr19:7712287 G>C, chr10:90771767 G>A, chr3:121415370 T>C, chr16:70503095 A>G, chr1:206945738 C>T, chr5:156593120 C>T, chr4:27019452 C>T, chr1:155317682 C>T, chr17:77926526 C>T, chr1:235840495 G>T, chr14:21993359 G>A, chr8:61757805 C>T, chr15:91306241 G>A, chr16:50741791 C>T, chr22:23915583 T>C, chr2:47205921 C>T, chr12:88900891 C>A, chr3:142281353 C>G, chr11:108123551 C>T, chr1:207641950 C>T, chr6:143092151 T>C, chr2:24431184 C>T, chr2:24432937 C>T, chr9:312134 G>A, chr8:100205255 G>A, chr21:16339852 T>C, and any combination thereof (see Tables 14 and 15).


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr16:81942175 A>G, chr2:163136505 C>G, chr11:67818269 G>A, chr22:23917192 G>190, chr20:3846397 C>T, chr8:145154222, G>A chr8:61654298 T>A, chr3:39323163 A>C, chr4:151199080 G>A, chr1:42047208 C>G, chr2:163124051 C>T, chr1:182554557 C>T, and any combination thereof (see Table 14).


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of chr8:145154824 A>C, chr20:62305450 C>T, chr22:23915745 G>A, chr6:83884161 C>G, chr11:108202772 G>T, chr5:138856923 C>T, chr16:1510535 C>T, chr20:3843027 C>A, chr12:122064788 G>GT, chr16:7714909 C>T, chr18:56401523 C>T, chr1:92946625 G>C, chr5:169081453 G>C, chr11:108117787 C>T, chr22:21235389 A>G, chr19:4817657 C>T, chr10:1060218 G>A, chr21:30698953 T>G, chr9:304628 G>A, chr19:7712287 G>C, chr10:90771767 G>A, chr3:121415370 T>C, chr16:70503095 A>G, chr1:206945738 C>T, chr5:156593120 C>T, chr4:27019452 C>T, chr1:155317682 C>T, chr17:77926526 C>T, chr1:235840495 G>T, chr14:21993359 G>A, chr8:61757805 C>T, chr15:91306241 G>A, chr16:50741791 C>T, chr22:23915583 T>C, chr2:47205921 C>T, chr12:88900891 C>A, chr3:142281353 C>G, chr11:108123551 C>T, chr1:207641950 C>T, chr6:143092151 T>C, chr2:24431184 C>T, chr2:24432937 C>T, chr9:312134 G>A, chr8:100205255 G>A, chr21:16339852 T>C, and any combination thereof (see Table 15).


In some embodiments, the SNV is a heterozygous SNV.


In some embodiments, the SNV is a homozygous SNV.


In some embodiments, the one or more genetic variations comprise a pair of single nucleotide variations (SNVs), wherein the pair of SNVs are encoded by any one of SEQ ID NO pairs: 1003 and 1004, 1003 and 1005, 1006 and 1007, 1024 and 1025, 1030 and 1031, 1047 and 1048, 1049 and 1050, 1063 and 1064, 1063 and 1065, 1063 and 1066, 1075 and 1076, 1091 and 1093, 1091 and 1096, 1093 and 1095, 1094 and 1097, 1098 and 1099, 1098 and 1100, 1099 and 1100, 1102 and 1103, 1104 and 1106, 1104 and 1107, 1104 and 1108, 1104 and 1109, 1104 and 1110, 1104 and 1111, 1104 and 1112, 1110 and 1111, 1112 and 1113, 1119 and 1120, 1124 and 1125, 1124 and 1126, 1125 and 1126, 1140 and 1141, 1142 and 1144, 1146 and 1151, 1147 and 1148, 1147 and 1149, 1153 and 1146, 1153 and 1147, 1155 and 1156, 1160 and 1161, 1165 and 1166, 1186 and 1187, 1188 and 1193, 1189 and 1193, 1191 and 1192, 1191 and 1193, 1191 and 1195, 1192 and 1193, 1192 and 1195, 1196 and 1197, 1206 and 1207, 1210 and 1218, 1211 and 1213, 1212 and 1213, 1213 and 1215, 1213 and 1216, 1213 and 1217, 1233 and 1238, 1242 and 1243, 1245 and 1246, 1263 and 1260, 1269 and 1279, 1270 and 1279, 1270 and 1282, 1271 and 1279, 1274 and 1279, 1278 and 1279, 1278 and 1281, 1279 and 1280, 1279 and 1281, 1279 and 1282, 1292 and 1293, 1296 and 1297, 1305 and 1314, 1306 and 1310, 1313 and 1321 or 1315 and 1322 (see Table 9 or Tables 9 and 7 for a subset), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 157, 2, 140, 65, 26, 14 or 45 (see Tables 7 and 8), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 2, 140, 65, 26, 14 or 45 (see Table 7), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to SEQ ID NO 157 (see Table 8), or a complement thereof


In some embodiments, the one or more genetic variations comprise a CNV and a single nucleotide variations (SNV), wherein SNVs is encoded by any one of SEQ ID NOs 1301, 1173, 1107, 1104, 1199, 1225, 1086 or 1223 (see Table 10), or complements thereof.


In some embodiments, the one or more genetic variations comprise a genetic variation selected from the group consisting of one or more of the following: chr8:145154222 G>A, chr2:163136505 C>G, chr16:81942175 A>G, and chr8:61654298 T>A (see Tables 14 and 16).


In some embodiments, the one or more genetic variations disrupt or modulate one or more of the following genes: PLCG2, POLE, LRBA, EPG5 and SHARPIN (see Table 17).


In some embodiments, the one or more genetic variations disrupt or modulate one or more of the following genes: PLCG2, CHD7, IFIH1, AP3B1, EPG5, PIK3CD, LRBA and SHARPIN (see Table 18).


In some embodiments, the gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 173-455 or 1500-2177 (see Tables 4 and 12), or complements thereof.


In some embodiments, the gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 173-455 (see Table 4), or complements thereof.


In some embodiments, the gene encodes a transcript with a sequence that has at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% sequence identity to any one of SEQ ID NOs 1500-2177 (see Table 12), or complements thereof.


In some embodiments, the one or more genetic variations comprise at least 5, at least 10, at least 20, or at least 50 genetic variations.


In some embodiments, panel of polynucleic acids comprises at least 5, at least 10, at least 20, or at least 50 polynucleic acids.


In some embodiments, the gene comprises a gene selected from the group consisting of gene numbers (GNs) 1-156 (in Table 3).


In some embodiments, the gene comprises a gene selected from the group consisting of gene numbers (GNs) in Table 6.


In some embodiments, the gene comprises a gene selected from the group consisting of PLCG2, RBCK1, EPG5, IL17F, SHARPIN, PRF1, JAGN1, TAP1, POLE, LRBA, EHF, IL12B, ATL2, NHEJ1, LYST, HIVEP1, AP3B1, TNFRSF10A, PIK3CD, PNP, MCEE, DOCK2 and ALG12 (see Table 13).


Provided herein is a method to predict an adverse responsiveness of a subject to a therapy, the method comprising detecting one or more genetic variations that disrupt or modulate a gene of GN1-GN490 in a polynucleic acid sample from the subject; and using that detection as a biomarker for predicting a response of the subject to the therapy to be adverse, wherein the therapy is an immunosuppressive therapy.


Provided herein is a method of screening for a PML biomarker comprising obtaining biological samples from subjects with PML; screening the biological samples to obtain nucleic acid information; detecting one or more genetic variations that disrupt or modulate a gene of GN1-GN490 in a polynucleic acid sample from a subject suspected of having PML; and using that detection as a biomarker for predicting a response of the subject to the therapy to be adverse, wherein the therapy is an immunosuppressive therapy.


Provided herein is a method of screening for a PML biomarker comprising obtaining biological samples from subjects with PML; screening the biological samples to obtain nucleic acid information; confirming each biological sample is not a duplicate of any other biological sample based on the nucleic acid information; detecting one or more genetic variations that disrupt or modulate a gene of GN1-GN490 in a polynucleic acid sample from a subject suspected of having PML; and using that detection as a biomarker for predicting a response of the subject to the therapy to be adverse, wherein the therapy is an immunosuppressive therapy.


Provided herein is a method of screening for a PML biomarker comprising obtaining biological samples from subjects with PML; screening the biological samples to obtain nucleic acid information; determining a sex genotype for each biological sample based on the nucleic acid information; confirming the sex genotype of each sample is the same as a sex phenotype of the subject from the subjects with PML; detecting one or more genetic variations that disrupt or modulate a gene of GN1-GN490 in a polynucleic acid sample from a subject suspected of having PML; and using that detection as a biomarker for predicting a response of the subject to the therapy to be adverse, wherein the therapy is an immunosuppressive therapy.


Provided herein is a method of treating a condition in a subject in need of natalizumab therapy, comprising: administering a therapeutically effective amount of natalizumab to the subject, wherein the subject has a decreased risk of progressive multifocal leukoencephalopathy (PML) due to an infection of the brain by John Cunningham virus (JCV), wherein the subject's decreased risk is due to the absence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


In some embodiments, the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, the subject is known as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, the subject is identified in a report (e.g., health report) as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


In some embodiments, the condition is multiple sclerosis or Crohn's disease. In some embodiments, the condition is a relapsing form of multiple sclerosis. In some embodiments, the natalizumab is administered via intravenous infusion.


In some embodiments, about 100 mg to about 500 mg of the natalizumab is administered. In some embodiments, about 100 mg to about 500 mg of the natalizumab is administered, for example, about 100 mg to about 200 mg, about 100 mg to about 300 mg, about 100 mg to about 400 mg, about 100 mg to about 500 mg, about 200 mg to about 300 mg, about 200 mg to about 400 mg, about 200 mg to about 500 mg, about 300 mg to about 400 mg, about 300 mg to about 500 mg, or about 400 mg to about 500 mg of the natalizumab is administered. In some embodiments, about 100 mg to about 500 mg of the natalizumab is administered via intravenous infusion. In some embodiments, about 100 mg to about 500 mg of the natalizumab is administered via intravenous infusion in four weeks. In some embodiments, about 300 mg of the natalizumab is administered. In some embodiments, about 300 mg of the natalizumab is administered via intravenous infusion. In some embodiments, about 300 mg of the natalizumab is administered via intravenous infusion in four weeks.


In some embodiments, the one or more genetic variations are associated with a risk of developing PML in a polynucleic acid sample from the subject. In some embodiments, the one or more genetic variations comprises a first genetic variation and a second genetic variation, wherein the first genetic variation disrupts or modulates a corresponding gene according to Tables 3 and 6, and wherein the second genetic variation disrupts or modulates a corresponding gene according to Tables 25A, 25B, and 26.


In some embodiments, the method comprises testing the subject for a genetic predisposition for PML with a genetic assay. In some embodiments, the genetic assay has a diagnostic yield of at least 5%. In some cases, the genetic assay has a diagnostic yield of at least about 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99%. In some cases, the genetic assay has a diagnostic yield of about 1%-5%, 1%-10%, 1%-20%, 5%-10%, 5%-20%, 10%-20%, 10%-30%, 20%-30%, 20%-40%, 30%-40%, 30%-50%, 40%-50%, 40%-60%, 50%-60%, 50%-70%, 60%-70%, 60%-80%, 70%-80%, 70%-90%, 80%-90%, 80%-95%, 90%-95%, 90%-99%, 90%-100%, 95%-99%, or 99%-100%. In some embodiments, the genetic assay has a diagnostic yield of at least 20%.


In some embodiments, the one or more genetic variations disrupt or modulate a corresponding gene according to Tables 13-18. In some embodiments, the one or more genetic variations disrupt or modulate a corresponding gene according to Tables 19-24.


In some embodiments, the subject's decreased risk is further due to the absence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 25A, 25B, and 26.


In some embodiments, the one or more genetic variations disrupt or modulate a corresponding gene selected from the group consisting of Homo sapiens chromodomain helicase DNA binding protein 7 (CHD7), Homo sapiens interferon induced with helicase C domain 1 (IFIH1), Homo sapiens immunoglobulin lambda like polypeptide 1 (IGLL1), Homo sapiens mitochondrial antiviral signaling protein (MAVS), Homo sapiens phospholipase C gamma 2 (PLCG2), Homo sapiens SHANK-associated RH domain interactor (SHARPIN), Homo sapiens T-cell immune regulator 1, ATPase H+ transporting VO subunit a3 (TCIRG1), and any combination thereof. In some embodiments, the one or more genetic variations comprise chr8:61654298 T>A, chr2:163136505 C>G, chr22:23917192 G>T, chr20:3846397 C>T, chr16:81942175 A>G, chr8:145154222 G>A, chr11:67818269 G>A, chr8:145154824 A>C, chr22:23915745 G>A, chr20:3843027 C>A, or any combination thereof.


In some embodiments, the corresponding gene comprises a gene selected from the group consisting of gene numbers (GNs) GN1-GN490. In some embodiments, the corresponding gene comprises a gene selected from the group consisting of gene numbers (GNs) GN1-GN241, GN243-GN369, and GN371-GN490.


In some embodiments, the one or more genetic variations are encoded by a sequence with at least 60% sequence identity to SEQ ID NOs 1-172 or SRN1-SRN363, with 100% sequence identity to SEQ ID NOs 1000-1329, or with at least 80% and less than 100% sequence identity to GN1-GN490, or complements thereof. In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV with at least 60% sequence identity to SEQ ID NOs 1-172, or complements thereof. In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a CNV sub-region (SRN) with at least 60% sequence identity to SRN1-SRN363, or complements thereof. In some embodiments, the one or more genetic variations comprise a genetic variation encoded by a single nucleotide variation (SNV) with a sequence of any one of SEQ ID NOs: 1000-1329, or complements thereof. In some embodiments, the one or more genetic variations are encoded by a sequence with at least 40% sequence identity to SEQ ID NOs 1-172 or SRN1-SRN363, for example, at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs 1-172 or SRN1-SRN363, or complements thereof. In some embodiments, the one or more genetic variations are encoded by a sequence with at least 40% sequence identity to SEQ ID NOs 1000-1329, for example, at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to SEQ ID NOs 1000-1329, or complements thereof. In some embodiments, the one or more genetic variations are encoded by a sequence with at least 40% and less than 100% sequence identity to GN1-GN490, for example, at least 40% and less than 50%, at least 50% and less than 60%, at least 60% and less than 70%, at least 70% and less than 80%, at least 80% and less than 90%, or at least 90% and less than 100% sequence identity to GN1-GN490, or complements thereof.


In some embodiments, the genetic assay comprises microarray analysis, PCR, sequencing, nucleic acid hybridization, or any combination thereof.


In some embodiments, the method comprises testing the subject with a JCV-antibody test, a CD62L test, or a CSF IgM oligoclonal bands test. In some embodiments, the method comprises testing the subject with the JCV-antibody test, wherein the JCV-antibody test does not detect a presence of JCV. In some embodiments, the method comprises testing the subject with the JCV-antibody test, wherein the JCV-antibody test detects a presence of JCV. In some embodiments, the JCV-antibody test comprises contacting a JCV detection reagent to a biological sample from the subject. In some embodiments, the JCV detection reagent is selected from the group consisting of an anti-JCV antibody, a JCV specific primer, and combinations thereof.


In some embodiments, the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a kit, comprising reagents for assaying a polynucleic acid sample from a subject in need thereof for the presence of one or more genetic variations that disrupt or modulate a gene of GN1-GN490. In some embodiments, the one or more genetic variations that disrupt or modulate a gene of GN1-GN241, GN243-GN369, and GN371-GN490.


Provided herein is a method of treating multiple sclerosis or Crohn's disease comprising: (a) testing a subject with multiple sclerosis or Crohn's disease for a genetic predisposition for PML with a genetic assay, wherein the genetic assay has a diagnostic yield of at least 20%, and (b) administering a therapeutically effective amount of natalizumab to the subject, wherein the testing does not identify the subject as having the genetic predisposition for PML.


In some embodiments, the method further comprises testing the subject with a JCV-antibody test. In some embodiments, the JCV-antibody test does not detect a presence of JCV. In some embodiments, the JCV-antibody test detects a presence of JCV. In some embodiments, the genetic assay tests the subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6.


Provided herein is a method of identifying a subject as not having a risk of developing PML, comprising: (a) analyzing a polynucleic acid sample from the subject for one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, wherein a genetic variation of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6 is not present in the polynucleic acid sample; and (b) identifying the subject as not having a risk of developing PML.


DETAILED DESCRIPTION OF THE DISCLOSURE

The details of one or more inventive embodiments are set forth in the accompanying drawings, the claims, and in the description herein. Other features, objects, and advantages of inventive embodiments disclosed and contemplated herein will be apparent from the description and drawings, and from the claims. As used herein, unless otherwise indicated, the article “a” means one or more unless explicitly otherwise provided for. As used herein, unless otherwise indicated, terms such as “contain,” “containing,” “include,” “including,” and the like mean “comprising.” As used herein, unless otherwise indicated, the term “or” can be conjunctive or disjunctive. As used herein, unless otherwise indicated, any embodiment can be combined with any other embodiment. As used herein, unless otherwise indicated, some inventive embodiments herein contemplate numerical ranges. When ranges are present, the ranges include the range endpoints. Additionally, every subrange and value within the range is present as if explicitly written out. The term “about” and its grammatical equivalents in relation to a reference numerical value and its grammatical equivalents as used herein can include a range of values plus or minus 10% from that value, such as a range of values plus or minus 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% from that value. For example, the amount “about 10” includes amounts from 9 to 11.


Progressive Multifocal Leukoencephalopathy (PML)


Progressive multifocal leukoencephalopathy (PML) is a rare and usually fatal viral disease characterized by progressive damage or inflammation of the white matter of the brain at multiple locations. The cause of PML can be a type of polyomavirus called the John Cunningham (JC) virus (or JCV), which can be harmless except in cases of weakened immune systems. While JCV is present at very high rates in the general population, PML remains a rare disorder, albeit an important one because of the clinical sequelae.


PML can occur in patients with severe immune deficiency, which allows reactivation of the JC virus, such as: 1) most commonly among patients with acquired immune deficiency syndrome (AIDS) that results from infection with human immunodeficiency virus (HIV), 2) patients on immunosuppressive medications like corticosteroids for organ transplant (e.g., renal, liver, lung, and heart) and in people with cancer (e.g., Hodgkin's disease, leukemia, or lymphoma, and myeloproliferative neoplasms such as myelofibrosis), and 3) individuals with autoimmune diseases (e.g., multiple sclerosis, rheumatoid arthritis, psoriasis, and systemic lupus erythematosus) with therapies that depress the immune response. Several immunosuppressive drugs have been reported in the context of drug-induced PML or drug-associated PML. For example, see: Melis et al. CNS Drugs. 2015; 29(10):879-91); Maas et al. J Neurol. 2016 October; 263(10):2004-21; Colin et al. Fundam Chin Pharmacol. 2016 Oct. 13. Immunosuppressive medications can include, but are not limited to, interferon beta-1a, interferon beta-1b, glatiramer acetate, peginterferon beta-1a, teriflunomide, mitoxantrone, ocrelizumab, abatacept, adalimumab, alefacept, alemtuzumab, anakinra, bortezomib (e.g., Velcade), eculizumab (e.g., Soliris), leflunomide, and various other transplant drugs such as antithymocyte Ig, asparaginase, azathioprine, basiliximab, belatacept, belimumab, bendamustine, bevacizumab, bleomycin, brentuximab vedotin, busulfan, capecitabine, carboplatin, carmustine, certolizumab, cetuximab, chlorambucil, cladribine, corticosteroids, cyclophosphamide, cyclosporine, cytotoxic chemotherapy, daclizumab, dimethyl fumarate, doxorubicin, efalizumab, etanercept, etoposide, fingolimod, fludarabine, gemcitabine, hydrocortisone, ibritumomab tiuxetan, ibrutinib, idarubicin, idelalisib, ifosfamide, imatinib, infliximab, lenalidomide, levamisole, mercaptopurine, methotrexate, mizoribine, muromonab-CD3, mycophenolate mofetil, mycophenolic acid, natalizumab, obinutuzumab, ofatumumab, oxaliplatin, prednisone/prednisolone, rituximab, ruxolitinib, sirolimus (also known as rapamycin), tacrolimus, tegafur/gimeracil/oteracil, thiotepa, tocilizumab, tofacitinib, ustekinumab, vedolizumab, vinblastine and vincristine. Exemplary small molecule immunosuppressive medications include dimethyl fumarate, fingolimod, and ruxolitinib. In some embodiments, an immunosuppressive therapy is classified as a Class 1 (high risk) therapeutic agent, such as efalizumab and natalizumab as reported in Calabrese L. H. et al., Nat Rev Rheumatol. (2015).


PML can be diagnosed in a patient with a progressive course of the disease, finding JC virus DNA in spinal fluid together with consistent white matter lesions on brain magnetic resonance imaging (MRI); alternatively, a brain biopsy can be diagnostic when the typical histopathology of demyelination, bizarre astrocytes, and enlarged oligodendroglial nuclei are present, coupled with techniques showing the presence of JC virus. Characteristic evidence of PML on brain CT scan images can be multifocal, non-contrast enhancing hypodense lesions without mass effect, but MRI can be more sensitive than CT. The most common area of involvement can be the cortical white matter of frontal and parieto-occipital lobes, but lesions may occur anywhere in the brain, like the basal ganglia, external capsule, and posterior cranial fossa structures like the brainstem and cerebellum.


In general, treatment of PML aims at reversing the immune deficiency to slow or stop the disease progress. Patients on an immunosuppression regime can stop taking the immunosuppressive medication or plasma exchange (PLEX) can be used to accelerate the removal of the immunosuppressive medication that put the person at risk for PML. HIV-infected patients can start highly active antiretroviral therapy (HAART). Occurrence of PML can also occur in the context of immune reconstitution inflammatory syndrome (IRIS), wherein onset of PML can occur or PML symptoms may get worse after cessation of immunosuppression (e.g., as reviewed by Pavlovic et al. Ther Adv Neurol Disord. 2015 November; 8(6):255-73 and Bowen et al. Nat Rev Neurol. 2016 Oct. 27; 12(11):662-674). For example, in MS patients that develop PML during treatment with natalizumab, IRIS often results when treatment is stopped and PLEX is used to remove natalizumab from the patient's circulation. Treatment of IRIS in PML patients can include administration of corticosteroids. Other potential treatments of PML can include cidofovir, cytarabine, anti-malaria drug mefloquine, interleukin-2, and 1-O-hexadecyloxypropyl-cidofovir (CMX001, aka brincidofovir). As reviewed by Pavlovic (Ther Adv Neurol Disord. 2015 November; 8(6):255-73), potential treatments for PML include antiviral agents (e.g., chlorpromazine, citalopram, mirtazapine, risperidone, ziprasidone, retro-2cycl, brefeldin A, cidofovir, brincidofovir, cytarabine, ganciclovir, leflunomide, topotecan, mefloquine, 3-aminobenzamide, imatinib, and Ag122), immune response modulators (e.g., IFN-alpha, IL-2, IL-7, maraviroc, and glucocorticoids), and immunization (e.g., recombinant human anti-JCV VP-1 monoclonal antibodies, JCV-specific cytotoxic T lymphocyte therapy, IL-7 plus JCV VP1 vaccine, and JCV oral vaccine).


The term “diagnostic yield” as used herein refers to the percentage of cases that would identify the presence of one or more genetic variations (e.g., CNV, SNV) in a PML cohort using an assay. For example, if 40 cases would identify the presence of one or more genetic variations (e.g., CNV, SNV) in a cohort of 100 PML patients, the diagnostic yield of the assay is 40%. In some cases, the patients in the PML cohort are clinically diagnosed with PML. In some cases, a patient is clinically diagnosed with PML when JC virus DNA is present in spinal fluid and consistent white matter lesions is present on brain magnetic resonance imaging (MRI). In some cases, a patient is clinically diagnosed with PML when typical histopathology of demyelination, bizarre astrocytes, and enlarged oligodendroglial nuclei are present in a brain biopsy, coupled with the presence of JC virus. In some cases, the PML cohort has at least 5 PML cases, for example, at least 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 PML cases. In some cases, the PML cohort is a cohort listed herein. For example, the PML cohort is the PML patient cohort listed in Table 7. In some cases, the assay is JCV-antibody assay. In some cases, the assay is not JCV-antibody assay. In some cases, the assay is a genetic assay. In some cases, the genetic assay tests the genetic predisposition for PML.


The genetic assay can comprise any method disclosed herein. In some cases, the genetic assay has a diagnostic yield of at least about 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99%. In some cases, the genetic assay has a diagnostic yield of about 1%-5%, 1%-10%, 1%-20%, 5%-10%, 5%-20%, 10%-20%, 10%-30%, 20%-30%, 20%-40%, 30%-40%, 30%-50%, 40%-50%, 40%-60%, 50%-60%, 50%-70%, 60%-70%, 60%-80%, 70%-80%, 70%-90%, 80%-90%, 80%-95%, 90%-95%, 90%-99%, 90%-100%, 95%-99%, or 99%-100%.


Genetic Variations Associated with PML


Described herein, are methods that can be used to detect genetic variations. Detecting specific genetic variations, for example polymorphic markers and/or haplotypes, copy number, absence or presence of an allele, or genotype associated with a condition (e.g., disease or disorder) as described herein, can be accomplished by methods known in the art for analyzing nucleic acids and/or detecting sequences at polymorphic or genetically variable sites, for example, amplification techniques, hybridization techniques, sequencing, microarrays/arrays, or any combination thereof. Thus, by use of these methods disclosed herein or other methods available to the person skilled in the art, one or more alleles at polymorphic markers, including microsatellites, single nucleotide polymorphisms (SNPs), single nucleotide variations (SNVs), insertions/deletions (indels), copy number variations (CNVs), or other types of genetic variations, can be identified in a sample obtained from a subject.


Genomic sequences within populations exhibit variability between individuals at many locations in the genome. For example, the human genome exhibits sequence variations that occur on average every 500 base pairs. Such genetic variations in polynucleic acid sequences are commonly referred to as polymorphisms or polymorphic sites. As used herein, a polymorphism, e.g., genetic variation, includes a variation in the sequence of the genome amongst a population, such as allelic variations and other variations that arise or are observed. Thus, a polymorphism refers to the occurrence of two or more genetically determined alternative sequences or alleles in a population. These differences can occur in coding (e.g., exonic) and non-coding (e.g., intronic or intergenic) portions of the genome, and can be manifested or detected as differences in polynucleic acid sequences, gene expression, including, for example transcription, processing, translation, transport, protein processing, trafficking, DNA synthesis; expressed proteins, other gene products or products of biochemical pathways or in post-translational modifications and any other differences manifested amongst members of a population. Polymorphisms that arise as the result of a single base change, such as single nucleotide polymorphisms (SNPs) or single nucleotide variations (SNVs), can include an insertion, deletion or change in one nucleotide. A polymorphic marker or site is the locus at which divergence occurs. Such sites can be as small as one base pair (an SNP or SNV). Polymorphic markers include, but are not limited to, restriction fragment length polymorphisms (RFLPs), variable number of tandem repeats (VNTRs), hypervariable regions, minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats and other repeating patterns, simple sequence repeats and insertional elements, such as Alu. Polymorphic forms also are manifested as different mendelian alleles for a gene. Polymorphisms can be observed by differences in proteins, protein modifications, RNA expression modification, DNA and RNA methylation, regulatory factors that alter gene expression and DNA replication, and any other manifestation of alterations in genomic polynucleic acid or organelle polynucleic acids. Those skilled in the art can appreciate that polymorphisms are sometimes considered to be a subclass of variations, defined on the basis of a particular frequency cutoff in a population. For example, in some embodiments, polymorphisms are considered to genetic variants/variations that occur at >1%, or >5%, frequency in the population.


In some embodiments, these genetic variations can be found to be associated with one or more disorders and/or diseases using the methods disclosed herein. In some embodiments, these genetic variations can be found to be associated with absence of one or more disorders and/or diseases (i.e. the one or more variants are protective against development of the disorder and/or diseases) using the methods disclosed herein.


In some embodiments, these genetic variations comprise point mutations, polymorphisms, single nucleotide polymorphisms (SNPs), single nucleotide variations (SNVs), translocations, insertions, deletions, amplifications, inversions, interstitial deletions, copy number variations (CNVs), loss of heterozygosity, or any combination thereof. As genetic variation includes any deletion, insertion or base substitution of the genomic DNA of one or more individuals in a first portion of a total population which thereby results in a difference at the site of the deletion, insertion or base substitution relative to one or more individuals in a second portion of the total population. Thus, the term “genetic variation” encompasses “wild type” or the most frequently occurring variation, and also includes “mutant,” or the less frequently occurring variation. In some embodiments, a wild type allele may be referred to as an ancestral allele.


As used herein, a target molecule that is “associated with” or “correlates with” a particular genetic variation is a molecule that can be functionally distinguished in its structure, activity, concentration, compartmentalization, degradation, secretion, and the like, as a result of such genetic variation. In some embodiments polymorphisms (e.g., polymorphic markers, genetic variations, or genetic variants) can comprise any nucleotide position at which two or more sequences are possible in a subject population. In some embodiments, each version of a nucleotide sequence, with respect to the polymorphism/variation, can represent a specific allele of the polymorphism/variation. In some embodiments, genomic DNA from a subject can contain two alleles for any given polymorphic marker, representative of each copy of the marker on each chromosome. In some embodiments, an allele can be a nucleotide sequence of a given location on a chromosome. Polymorphisms/variations can comprise any number of specific alleles. In some embodiments of the disclosure, a polymorphism/variation can be characterized by the presence of two or more alleles in a population. In some embodiments, the polymorphism/variation can be characterized by the presence of three or more alleles. In some embodiments, the polymorphism/variation can be characterized by four or more alleles, five or more alleles, six or more alleles, seven or more alleles, nine or more alleles, or ten or more alleles. In some embodiments an allele can be associated with one or more diseases or disorders, for example, a PML risk allele can be an allele that is associated with increased or decreased risk of developing PML. In some embodiments, genetic variations and alleles can be used to associate an inherited phenotype with a responsible genotype. In some embodiments, a PML risk allele can be a variant allele that is statistically associated with a screening of PML. In some embodiments, genetic variations can be of any measurable frequency in the population, for example, a frequency higher than 10%, a frequency from 5-10%, a frequency from 1-5%, a frequency from 0.1-1%, or a frequency below 0.1%. As used herein, variant alleles can be alleles that differ from a reference allele. As used herein, a variant can be a segment of DNA that differs from the reference DNA, such as a genetic variation. In some embodiments, genetic variations can be used to track the inheritance of a gene that has not yet been identified, but whose approximate location is known.


As used herein, a “haplotype” can be information regarding the presence or absence of one or more genetic markers in a given chromosomal region in a subject. In some embodiments, a haplotype can be a segment of DNA characterized by one or more alleles arranged along the segment, for example, a haplotype can comprise one member of the pair of alleles for each genetic variation or locus. In some embodiments, the haplotype can comprise two or more alleles, three or more alleles, four or more alleles, five or more alleles, or any combination thereof, wherein, each allele can comprise one or more genetic variations along the segment.


In some embodiments, a genetic variation can be a functional aberration that can alter gene function, gene expression, polypeptide expression, polypeptide function, or any combination thereof. In some embodiments, a genetic variation can be a loss-of-function mutation, gain-of-function mutation, dominant negative mutation, or reversion. In some embodiments, a genetic variation can be part of a gene's coding region or regulatory region. Regulatory regions can control gene expression and thus polypeptide expression. In some embodiments, a regulatory region can be a segment of DNA wherein regulatory polypeptides, for example, transcription or splicing factors, can bind. In some embodiments a regulatory region can be positioned near the gene being regulated, for example, positions upstream or downstream of the gene being regulated. In some embodiments, a regulatory region (e.g., enhancer element) can be several thousands of base pairs upstream or downstream of a gene.


In some embodiments, variants can include changes that affect a polypeptide, such as a change in expression level, sequence, function, localization, binding partners, or any combination thereof. In some embodiments, a genetic variation can be a frameshift mutation, nonsense mutation, missense mutation, neutral mutation, or silent mutation. For example, sequence differences, when compared to a reference nucleotide sequence, can include the insertion or deletion of a single nucleotide, or of more than one nucleotide, resulting in a frame shift; the change of at least one nucleotide, resulting in a change in the encoded amino acid; the change of at least one nucleotide, resulting in the generation of a premature stop codon; the deletion of several nucleotides, resulting in a deletion of one or more amino acids encoded by the nucleotides; the insertion of one or several nucleotides, such as by unequal recombination or gene conversion, resulting in an interruption of the coding sequence of a reading frame; duplication of all or a part of a sequence; transposition; or a rearrangement of a nucleotide sequence. Such sequence changes can alter the polypeptide encoded by the nucleic acid, for example, if the change in the nucleic acid sequence causes a frame shift, the frame shift can result in a change in the encoded amino acids, and/or can result in the generation of a premature stop codon, causing generation of a truncated polypeptide. In some embodiments, a genetic variation associated with PML can be a synonymous change in one or more nucleotides, for example, a change that does not result in a change in the amino acid sequence. Such a polymorphism can, for example, alter splice sites, affect the stability or transport of mRNA, or otherwise affect the transcription or translation of an encoded polypeptide. In some embodiments, a synonymous mutation can result in the polypeptide product having an altered structure due to rare codon usage that impacts polypeptide folding during translation, which in some cases may alter its function and/or drug binding properties if it is a drug target. In some embodiments, the changes that can alter DNA increase the possibility that structural changes, such as amplifications or deletions, occur at the somatic level. A polypeptide encoded by the reference nucleotide sequence can be a reference polypeptide with a particular reference amino acid sequence, and polypeptides encoded by variant nucleotide sequences can be variant polypeptides with variant amino acid sequences.


The most common sequence variants comprise base variations at a single base position in the genome, and such sequence variants, or polymorphisms, are commonly called single nucleotide polymorphisms (SNPs) or single nucleotide variants (SNVs). In some embodiments, a SNP represents a genetic variant present at greater than or equal to 1% occurrence in a population and in some embodiments a SNP or an SNV can represent a genetic variant present at any frequency level in a population. A SNP can be a nucleotide sequence variation occurring when a single nucleotide at a location in the genome differs between members of a species or between paired chromosomes in a subject. SNPs can include variants of a single nucleotide, for example, at a given nucleotide position, some subjects can have a ‘G’, while others can have a ‘C’. SNPs can occur in a single mutational event, and therefore there can be two possible alleles possible at each SNP site; the original allele and the mutated allele. SNPs that are found to have two different bases in a single nucleotide position are referred to as biallelic SNPs, those with three are referred to as triallelic, and those with all four bases represented in the population are quadallelic. In some embodiments, SNPs can be considered neutral. In some embodiments SNPs can affect susceptibility to a condition (e.g., PML). SNP polymorphisms can have two alleles, for example, a subject can be homozygous for one allele of the polymorphism wherein both chromosomal copies of the individual have the same nucleotide at the SNP location, or a subject can be heterozygous wherein the two sister chromosomes of the subject contain different nucleotides. The SNP nomenclature as reported herein is the official Reference SNP (rs) ID identification tag as assigned to each unique SNP by the National Center for Biotechnological Information (NCBI).


Another genetic variation of the disclosure can be copy number variations (CNVs). As used herein, “CNVs” include alterations of the DNA of a genome that results in an abnormal number of copies of one or more sections of DNA. In some embodiments, a CNV comprises a CNV-subregion. As used herein, a “CNV-subregion” includes a continuous nucleotide sequence within a CNV. In some embodiments, the nucleotide sequence of a CNV-subregion can be shorter than the nucleotide sequence of the CNV, and in another embodiment the CNV-subregion can be equivalent to the CNV (e.g., such as for some recurrent CNVs). CNVs can be inherited or caused by de novo mutation and can be responsible for a substantial amount of human phenotypic variability, behavioral traits, and disease susceptibility. In some embodiments, CNVs of the current disclosure can be associated with susceptibility to one or more conditions, for example, PML. In some embodiments, CNVs can include a single gene or include a contiguous set of genes. In some embodiments, CNVs can be caused by structural rearrangements of the genome, for example, unbalanced translocations or inversions, insertions, deletions, amplifications, and interstitial deletions. In some embodiments, these structural rearrangements occur on one or more chromosomes. Low copy repeats (LCRs), which are region-specific repeat sequences (also known as segmental duplications), can be susceptible to these structural rearrangements, resulting in CNVs. Factors such as size, orientation, percentage similarity and the distance between the copies can influence the susceptibility of LCRs to genomic rearrangement. In addition, rearrangements may be mediated by the presence of high copy number repeats, such as long interspersed elements (LINES) and short interspersed elements (SINEs), often via non-homologous recombination. For example, chromosomal rearrangements can arise from non-allelic homologous recombination during meiosis or via a replication-based mechanism such as fork stalling and template switching (FoSTeS) (Zhang F. et al., Nat. Genet. (2009)) or microhomology-mediated break-induced repair (MMBIR) (Hastings P. J. et al., PLoS Genetics (2009)). In some embodiments, CNVs are referred to as structural variants, which are a broader class of variant that also includes copy number neutral alterations such as balanced inversions and balanced translocations.


CNVs can account for genetic variation affecting a substantial proportion of the human genome, for example, known CNVs can cover over 15% of the human genome sequence (Estivill and Armengol, PLoS Genetics (2007)). CNVs can affect gene expression, phenotypic variation and adaptation by disrupting or impairing gene dosage, and can cause disease, for example, microdeletion and microduplication disorders, and can confer susceptibility to diseases and disorders. Updated information about the location, type, and size of known CNVs can be found in one or more databases, for example, the Database of Genomic Variants (See, MacDonald J R et al., Nucleic Acids Res., 42, D986-92 (2014), which currently contains data for over 500,000 CNVs (as of May, 2016).


Other types of sequence variants can be found in the human genome and can be associated with a disease or disorder, including but not limited to, microsatellites. Microsatellite markers are stable, polymorphic, easily analyzed, and can occur regularly throughout the genome, making them especially suitable for genetic analysis. A polymorphic microsatellite can comprise multiple small repeats of bases, for example, CA repeats, at a particular site wherein the number of repeat lengths varies in a population. In some embodiments, microsatellites, for example, variable number of tandem repeats (VNTRs), can be short segments of DNA that have one or more repeated sequences, for example, about 2 to 5 nucleotides long, that can occur in non-coding DNA. In some embodiments, changes in microsatellites can occur during genetic recombination of sexual reproduction, increasing or decreasing the number of repeats found at an allele, or changing allele length.


The genetic variations disclosed herein can be associated with a risk of developing PML in a subject. In some cases, the subject can have a decreased risk due to the absence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 1 to 26. For example, the subject can have a decreased risk due to the absence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some cases, the subject can have an increased risk due to the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 1 to 26. For example, the subject can have an increased risk due to the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some cases, one or more genes listed in Tables 25A, 25B, and 26 can be removed from any one of the Tables 1-24. In some cases, one or more genes listed in Tables 25A, 25B, and 26 can be added to any one of the Tables 1-24.









TABLE 25A







exemplary 8-gene panel











RefSeq



Gene


Gene
Disease
Gene

Number


Symbol
Model
Source
Source Annotation
(GN)





BAG3
AR
Public_db
PMID: 19229298, 19282432, 22984599, 27042682
175


BTK
XLR
Public_db
PMID: 18281276, 23765059, 25930993, 26029204
180


CD40LG
XLR
Public_db
PMID: 17360404, 21455173, 23765059, 26008899,
206





26029204



DOCK8
AR
Public_db
PMID: 23765059, 23887241, 26029204, 26454313
242


MAGT1
XLR
Public_db
PMID: 23887241, 25504528, 27873163
326


RAG1
AD_AR
Public_db
PMID: 23122631, 23765059, 23887241, 25976673,
370





26029204, 26454313, 27484032, 27808398



STAT1
AD_AR
Public_db
PMID: 23887241, 25645939, 26029204, 26513235,
436





26743090, 27821552, 27873163



WAS
XLR
Both
PMID: 12874226, 14647476, 19782549, 20008220,
483





24753205, 26029204, 26371186
















TABLE 25B







exemplary 16-gene panel











RefSeq



Gene


Gene
Disease
Gene

Number


Symbol
Model
Source
Source Annotation
(GN)














ADA
AR
Both
PMID: 23765059, 24135998, 25930993, 26029204,
1





26454313



BAG3
AR
Public_db
PMID: 19229298, 19282432, 22984599, 27042682
175


BTK
XLR
Public_db
PMID: 18281276, 23765059, 25930993, 26029204
180


CD40LG
XLR
Public_db
PMID: 14647476, 17360404, 21455173, 23765059,
206





26008899, 26029204



DNMT3B
AR
Public_db
PMID: 23486536, 23765059, 26029204, 26851945
240


DOCK8
AR
Public_db
PMID: 23765059, 23887241, 26029204, 26454313
242


ITK
AR
Public_db
PMID: 14647476, 23765059, 26029204, 26454313
308


LCK
AR
Public_db
PMID: 14647476, 23765059, 26029204, 26454313
316


PNP
AR
Both
PMID: 26029204, 26454313
354


RAG1
AD_AR
Public_db
PMID: 23122631, 23765059, 23887241, 25976673,
370





26029204, 26454313, 27484032, 27808398



STAT1
AD_AR
Public_db
PMID: 23887241, 25645939, 26029204, 26513235,
436





26743090, 27821552, 27873163



STAT3
AD
Public_db
PMID: 23765059, 23887241, 25645939, 25930993,
438





26029204, 27658964, 27873163



STK3
unknown
Both
PMID: 26029204
135


TYK2
AR
Public_db
PMID: 26029204, 26513235, 27821552
144


WAS
XLR
Both
PMID: 12874226, 19782549, 20008220, 24753205,
483





26029204, 26371186



WIPF1
AR
Public_db
PMID: 23765059, 26029204, 26453379
485
















TABLE 26







exemplary 2-gene panel












RefSeq

NCBI


Gene


Gene
Exon
Gene
Gene

#


Symbol
overlap
ID
Description
RefSeq_Summary
(GN)















ADA
intronic
100
adenosine
This gene encodes an enzyme that catalyzes the
1





deaminase
hydrolysis of adenosine to inosine. Various mutations







have been described for this gene and have been linked







to human diseases. Deficiency in this enzyme causes a







form of severe combined immunodeficiency disease







(SCID), in which there is dysfunction of both B and T







lymphocytes with impaired cellular immunity and







decreased production of immunoglobulins, whereas







elevated levels of this enzyme have been associated with







congenital hemolytic anemia. [provided by RefSeq, July







2008]. Publication Note: This RefSeq record includes a







subset of the publications that are available for this







gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript







exon combination :: BC040226.1, X02994.1







[ECO:0000332] RNAseq introns :: mixed/partial sample







support ERS025081, ERS025082 [ECO:0000350]







##Evidence-Data-END##



STK3
intronic
6788
serine/
This gene encodes a serine/threonine protein kinase
135





threonine-
activated by proapoptotic molecules indicating the






protein
encoded protein functions as a growth suppressor.






kinase 3
Cleavage of the protein product by caspase removes the






isoform 1
inhibitory C-terminal portion. The N-terminal portion is







transported to the nucleus where it homodimerizes to







form the active kinase which promotes the condensation







of chromatin during apoptosis. Multiple transcript







variants encoding different isoforms have been found







for this gene. [provided by RefSeq, January 2012].







Transcript Variant: This variant (1) encodes isoform 1.







Publication Note: This RefSeq record includes a subset







of the publications that are available for this gene.







Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript







exon combination :: U26424.1, BC010640.2







[ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025084, ERS025088







[ECO:0000348] ##Evidence-Data-END##









A “subject”, as used herein, can be an individual of any age or sex from whom a sample containing polynucleotides is obtained for analysis by one or more methods described herein so as to obtain polynucleic acid information; for example, a male or female adult, child, newborn, or fetus. In some embodiments, a subject can be any target of therapeutic administration. In some embodiments, a subject can be a test subject or a reference subject.


As used herein, a “cohort” can represent an ethnic group, a patient group, a particular age group, a group not associated with a particular condition (e.g., disease or disorder), a group associated with a particular condition (e.g., disease or disorder), a group of asymptomatic subjects, a group of symptomatic subjects, or a group or subgroup of subjects associated with a particular response to a treatment regimen or enrolled in a clinical trial. In some embodiments, a patient can be a subject afflicted with a condition (e.g., disease or disorder). In some embodiments, a patient can be a subject not afflicted with a condition (e.g., disease or disorder) and is considered apparently healthy, or a normal or control subject. In some embodiments, a subject can be a test subject, a patient or a candidate for a therapeutic, wherein genomic DNA from the subject, patient, or candidate is obtained for analysis by one or more methods of the present disclosure herein, so as to obtain genetic variation information of the subject, patient or candidate.


In some embodiments, the polynucleic acid sample can be obtained prenatally from a fetus or embryo or from the mother, for example, from fetal or embryonic cells in the maternal circulation. In some embodiments, the polynucleic acid sample can be obtained with the assistance of a health care provider, for example, to draw blood. In some embodiments, the polynucleic acid sample can be obtained without the assistance of a health care provider, for example, where the polynucleic acid sample is obtained non-invasively, such as a saliva sample, or a sample comprising buccal cells that is obtained using a buccal swab or brush, or a mouthwash sample.


The present disclosure also provides methods for assessing genetic variations in subjects who are members of a target population. Such a target population is in some embodiments a population or group of subjects at risk of developing the condition (e.g., disease or disorder), based on, for example, other genetic factors, biomarkers, biophysical parameters, diagnostic testing such as magnetic resonance imaging (MRI), family history of the condition, previous screening or medical history, or any combination thereof.


The genetic variations of the present disclosure found to be associated with a condition (e.g., disease or disorder) can show similar association in other human populations. Particular embodiments comprising subject human populations are thus also contemplated and within the scope of the disclosure. Such embodiments relate to human subjects that are from one or more human populations including, but not limited to, Caucasian, Ashkenazi Jewish, Sephardi Jewish, European, American, Eurasian, Asian, Central/South Asian, East Asian, Middle Eastern, African, Hispanic, Caribbean, and Oceanic populations. European populations include, but are not limited to, Swedish, Norwegian, Finnish, Russian, Danish, Icelandic, Irish, Kelt, English, Scottish, Dutch, Belgian, French, German, Spanish, Portuguese, Italian, Polish, Bulgarian, Slavic, Serbian, Bosnian, Czech, Greek and Turkish populations. The ethnic contribution in subjects can also be determined by genetic analysis, for example, genetic analysis of ancestry can be carried out using unlinked microsatellite markers or single nucleotide polymorphisms (SNPs) such as those set out in Smith et al., (Smith M. W. et al., Am. J. Hum. Genet., 74:1001 (2004)).


Certain genetic variations can have different population frequencies in different populations, or are polymorphic in one population but not in another. The methods available and as thought herein can be applied to practice the present disclosure in any given human population. This can include assessment of genetic variations of the present disclosure, so as to identify those markers that give strongest association within the specific population. Thus, the at-risk variants of the present disclosure can reside on different haplotype background and in different frequencies in various human populations.


Conditions and Immunosuppressive Medications


In some embodiments, a subject can be diagnosed or undiagnosed with a condition (e.g., disease or disorder), can be asymptomatic or symptomatic, can have increased or decreased susceptibility to a condition (e.g., disease or disorder), can be currently under or previously under or not under a treatment for a condition (e.g., disease or disorder), or any combination thereof. In some embodiments, the condition can be AIDS, cancer, organ transplant, or an autoimmune disease. In some embodiments, the condition is PML.


In some embodiments, a subject can be diagnosed or undiagnosed with PML, can be asymptomatic or symptomatic, can have increased or decreased susceptibility to PML, can be currently under or previously under or not under a treatment for PML, or any combination thereof. In some embodiments, a subject can be diagnosed or undiagnosed with AIDS (e.g., individuals infected with HIV), can be asymptomatic or symptomatic, can have increased or decreased susceptibility to AIDS, can be currently under or previously under or not under a treatment for AIDS, or any combination thereof. In some embodiments, a subject can be diagnosed or undiagnosed with cancer (e.g., Hodgkin's disease, leukemia, lymphoma, or myelofibrosis), can be asymptomatic or symptomatic, can have increased or decreased susceptibility to cancer, can be currently under or previously under or not under a treatment for cancer, or any combination thereof. In some embodiments, a subject can be currently diagnosed or previously diagnosed or undiagnosed with an autoimmune disease (e.g., multiple sclerosis, rheumatoid arthritis, psoriasis, systemic lupus erythematosus), can be asymptomatic or symptomatic, can have increased or decreased susceptibility to an autoimmune disease, can be currently under or previously under or not under a treatment for an autoimmune disease, or any combination thereof.


The term “cancer” is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. A metastatic tumor can arise from a multitude of primary tumor types, including but not limited to those of breast, lung, liver, colon and ovarian origin. Examples of cancers include, but are not limited to, a fibrosarcoma, myosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, gastric cancer, esophageal cancer, rectal cancer, pancreatic cancer, ovarian cancer, prostate cancer, uterine cancer, cancer of the head and neck, skin cancer, brain cancer, squamous cell carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular cancer, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma, leukemia, lymphoma, myelofibrosis, or Kaposi sarcoma.


The term “autoimmune disease” is meant to include all types of pathological states arising from abnormal immune responses of the body to substances and tissues that are normally present in the body. Examples of autoimmune diseases include, but are not limited to, Addison disease, Anti-NMDA receptor encephalitis, antisynthetase syndrome, Aplastic anemia, autoimmune anemias, Autoimmune hemolytic anemia, Autoimmune pancreatitis, Behcet's Disease, bullous skin disorders, Celiac disease-sprue (gluten-sensitive enteropathy), chronic fatigue syndrome, Chronic inflammatory demyelinating polyneuropathy, chronic lymphocytic leukemia, Crohn's disease, Dermatomyositis, Devic's disease, Erythroblastopenia, Evans syndrome, Focal segmental glomerulosclerosis, Granulomatosis with polyangiitis, Graves disease, Graves' ophthalmopathy, Guillain-Barre syndrome, Hashimoto thyroiditis, idiopathic thrombocytopenic purpura (ITP), IgA nephropathy, IgA-mediated autoimmune diseases, IgG4-related disease, Inflammatory bowel disease, Juvenile idiopathic arthritis, Multiple sclerosis, Myasthenia gravis, myeloma, non-Hodgkin's lymphoma, Opsoclonus myoclonus syndrome (OMS), Pemphigoid, Pemphigus, pemphigus vulgaris, Pernicious anemia, polymyositis, Psoriasis, pure red cell aplasia, Reactive arthritis, Rheumatoid arthritis, Sarcoidosis, scleroderma, Sjögren syndrome, Systemic lupus erythematosus, Thrombocytopenic purpura, Thrombotic thrombocytopenic purpura, Type I diabetes, Ulcerative colitis, Vasculitis (e.g., vasculitis associated with anti-neutrophil cytoplasmic antibody) and Vitiligo.


In some embodiments, a subject can be currently treated with an immunosuppressive medication. In some embodiments, a subject can be previously treated with an immunosuppressive medication. In some embodiments, a subject can be not yet treated with an immunosuppressive medication. The immunosuppressive medication can include but not limited to glucocorticoids, cytostatics, antibodies, drugs acting on immunophilins, interferons, opioids, TNF binding proteins, mycophenolate, or other small biological agents. For example, glucocorticoids can include but not limited to cortisol (hydrocortisone), cortisone, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, beclometasone, fludrocortisone acetate, deoxycorticosterone acetate (DOCA), or aldosterone. Cytostatics can include but not limited to nitrogen mustards (cyclophosphamide), nitrosoureas, platinum compounds, folic acid analogues such as methotrexate, purine analogues such as azathioprine and mercaptopurine, pyrimidine analogues such as fluorouracil, protein synthesis inhibitors, cytotoxic antibiotics such as dactinomycin, anthracyclines, mitomycin C, bleomycin, or mithramycin. Antibodies can include but not limited to polyclonal antibodies such as atgam and thymoglobuline, monoclonal antibodies such as CD25- and CD3-directed antibodies, muromonab-CD3, basiliximab (Simulect), and daclizumab (Zenapax). Drugs acting on immunophilins can include but not limited to ciclosporin, tacrolimus, sirolimus, or everolimus. TNF binding proteins can include but not limited to infliximab (Remicade), etanercept (Enbrel), or adalimumab (Humira). Other small biological agents can include but not limited to fingolimod and myriocin.


In some embodiments, the immunosuppressive medication can be drugs for treating multiple sclerosis include but not limited to interferon beta-1a (e.g., Avonex, Rebif), interferon beta-1b (e.g., Betaseron, Extavia), glatiramer acetate (Copaxone, Glatopa), peginterferon beta-1a (e.g., Plegridy), teriflunomide (Aubagio), fingolimod (Gilenya), dimethyl fumarate (Tecfidera), alemtuzumab (Lemtrada), mitoxantrone (e.g., Novantrone), natalizumab (e.g., Tysabri), daclizumab (e.g., Zinbryta), or ocrelizumab (e.g., Ocrevus).


In some embodiments, the immunosuppressive medication can be adalimumab (e.g., Humira), alemtuzumab (e.g., Lemtrada), alentuzumab (e.g., Campath), azathioprine (e.g., Imuran), belimumab (e.g., Benlysta), bevacizumab (e.g., Avastatin), bortezomib (e.g., Velcade), eculizumab (e.g., Soliris), leflunomide, brentuximab vedotin (e.g., Adcetris), cetuximab (e.g., Erbitux), cyclophosphamid, cimethyl fumarate (e.g., Tecfidera), efalizumab (e.g., Raptiva), fingolimod (e.g., Gilenya), fludarabine (e.g., Fludara), fumaric acid, imatinib (e.g., Gleevec, Glivec), infliximab (e.g., Remicade), methotrexate (e.g., Trexall, Rheumatrex), mycophenolate mofetil (e.g., Cellcept), natalizumab (e.g., Tysabri), daclizumab (e.g., Zinbryta), rituximab (e.g., Rituxin), vedolizumab (Entyvio), ruxolitinib (e.g., Jakafi, Jakavi), or ocrelizumab (e.g., Ocrevus).


In some embodiments, a method of treating a condition in a subject in need of natalizumab therapy, comprises administering a therapeutically effective amount of natalizumab to the subject, wherein the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, a method of reducing a risk of a subject developing PML comprises administering a therapeutically effective amount of natalizumab to the subject, wherein the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, the condition is multiple sclerosis. In some embodiments, the condition is Crohn's disease. In some embodiments, a method of treating multiple sclerosis comprises administering natalizumab to a subject with multiple sclerosis, wherein the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, a method of treating Crohn's disease comprises administering natalizumab to a subject with Crohn's disease, wherein the subject is identified as not having one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, a method of treating multiple sclerosis comprises testing a subject with multiple sclerosis for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject does not have the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and administering natalizumab to the subject that was determined not to have the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, a method of treating Crohn's disease comprises testing a subject with Crohn's disease for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject does not have the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and administering natalizumab to the subject that was determined not to have the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, a method of reducing a risk of a subject developing PML comprises testing a subject for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject has at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and advising against administering natalizumab to the subject that was determined to have at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, the subject has multiple sclerosis. In some embodiments, the subject has Crohn's disease. In some embodiments, a method of treating multiple sclerosis comprises testing a subject with multiple sclerosis for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject has at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and advising against administering natalizumab to the subject that was determined to have at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, a method of treating Crohn's disease comprises testing a subject with Crohn's disease for the presence of one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, determining that the subject has at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6, and advising against administering natalizumab to the subject that was determined to have at least one of the one or more genetic variations that disrupt or modulate a corresponding gene according to Tables 3 and 6. In some embodiments, the advising comprises advising that administering natalizumab is contraindicated. In some embodiments, the advising comprises advising that administering natalizumab increases the risk of the subject developing PML. In some embodiments, the advising comprises advising that administering natalizumab is a factor that increases the risk of the subject developing PML.


Samples


Samples that are suitable for use in the methods described herein can be polynucleic acid samples from a subject. A “polynucleic acid sample” as used herein can include RNA or DNA, or a combination thereof. In another embodiment, a “polypeptide sample” (e.g., peptides or proteins, or fragments therefrom) can be used to ascertain information that an amino acid change has occurred, which is the result of a genetic variant. Polynucleic acids and polypeptides can be extracted from one or more samples including but not limited to, blood, saliva, urine, mucosal scrapings of the lining of the mouth, expectorant, serum, tears, skin, tissue, or hair. A polynucleic acid sample can be assayed for polynucleic acid information. “Polynucleic acid information,” as used herein, includes a polynucleic acid sequence itself, the presence/absence of genetic variation in the polynucleic acid sequence, a physical property which varies depending on the polynucleic acid sequence (e.g., Tm), and the amount of the polynucleic acid (e.g., number of mRNA copies). A “polynucleic acid” means any one of DNA, RNA, DNA including artificial nucleotides, or RNA including artificial nucleotides. As used herein, a “purified polynucleic acid” includes cDNAs, fragments of genomic polynucleic acids, polynucleic acids produced using the polymerase chain reaction (PCR), polynucleic acids formed by restriction enzyme treatment of genomic polynucleic acids, recombinant polynucleic acids, and chemically synthesized polynucleic acid molecules. A “recombinant” polynucleic acid molecule includes a polynucleic acid molecule made by an artificial combination of two otherwise separated segments of sequence, e.g., by chemical synthesis or by the manipulation of isolated segments of polynucleic acids by genetic engineering techniques. As used herein, a “polypeptide” includes proteins, fragments of proteins, and peptides, whether isolated from natural sources, produced by recombinant techniques, or chemically synthesized. A polypeptide may have one or more modifications, such as a post-translational modification (e.g., glycosylation, phosphorylation, etc.) or any other modification (e.g., pegylation, etc.). The polypeptide may contain one or more non-naturally-occurring amino acids (e.g., such as an amino acid with a side chain modification).


In some embodiments, the polynucleic acid sample can comprise cells or tissue, for example, cell lines. Exemplary cell types from which nucleic acids can be obtained using the methods described herein include, but are not limited to, the following: a blood cell such as a B lymphocyte, T lymphocyte, leukocyte, erythrocyte, macrophage, or neutrophil; a muscle cell such as a skeletal cell, smooth muscle cell or cardiac muscle cell; a germ cell, such as a sperm or egg; an epithelial cell; a connective tissue cell, such as an adipocyte, chondrocyte; fibroblast or osteoblast; a neuron; an astrocyte; a stromal cell; an organ specific cell, such as a kidney cell, pancreatic cell, liver cell, or a keratinocyte; a stem cell; or any cell that develops therefrom. A cell from which nucleic acids can be obtained can be a blood cell or a particular type of blood cell including, for example, a hematopoietic stem cell or a cell that arises from a hematopoietic stem cell such as a red blood cell, B lymphocyte, T lymphocyte, natural killer cell, neutrophil, basophil, eosinophil, monocyte, macrophage, or platelet. Generally, any type of stem cell can be used including, without limitation, an embryonic stem cell, adult stem cell, or pluripotent stem cell.


In some embodiments, a polynucleic acid sample can be processed for RNA or DNA isolation, for example, RNA or DNA in a cell or tissue sample can be separated from other components of the polynucleic acid sample. Cells can be harvested from a polynucleic acid sample using standard techniques, for example, by centrifuging a cell sample and resuspending the pelleted cells, for example, in a buffered solution, for example, phosphate-buffered saline (PBS). In some embodiments, after centrifuging the cell suspension to obtain a cell pellet, the cells can be lysed to extract DNA. In some embodiments, the nucleic acid sample can be concentrated and/or purified to isolate DNA. All nucleic acid samples obtained from a subject, including those subjected to any sort of further processing, are considered to be obtained from the subject. In some embodiments, standard techniques and kits known in the art can be used to extract RNA or DNA from a nucleic acid sample, including, for example, phenol extraction, a QIAamp® Tissue Kit (Qiagen, Chatsworth, Calif.), a Wizard® Genomic DNA purification kit (Promega), or a Qiagen Autopure method using Puregene chemistry, which can enable purification of highly stable DNA well-suited for archiving.


In some embodiments, determining the identity of an allele or determining copy number can, but need not, include obtaining a polynucleic acid sample comprising RNA and/or DNA from a subject, and/or assessing the identity, copy number, presence or absence of one or more genetic variations and their chromosomal locations within the genomic DNA (i.e. subject's genome) derived from the polynucleic acid sample.


The individual or organization that performs the determination need not actually carry out the physical analysis of a nucleic acid sample from a subject. In some embodiments, the methods can include using information obtained by analysis of the polynucleic acid sample by a third party. In some embodiments, the methods can include steps that occur at more than one site. For example, a polynucleic acid sample can be obtained from a subject at a first site, such as at a health care provider or at the subject's home in the case of a self-testing kit. The polynucleic acid sample can be analyzed at the same or a second site, for example, at a laboratory or other testing facility.


Nucleic Acids


The nucleic acids and polypeptides described herein can be used in methods and kits of the present disclosure. In some embodiments, aptamers that specifically bind the nucleic acids and polypeptides described herein can be used in methods and kits of the present disclosure. As used herein, a nucleic acid can comprise a deoxyribonucleotide (DNA) or ribonucleotide (RNA), whether singular or in polymers, naturally occurring or non-naturally occurring, double-stranded or single-stranded, coding, for example a translated gene, or non-coding, for example a regulatory region, or any fragments, derivatives, mimetics or complements thereof. In some embodiments, nucleic acids can comprise oligonucleotides, nucleotides, polynucleotides, nucleic acid sequences, genomic sequences, complementary DNA (cDNA), antisense nucleic acids, DNA regions, probes, primers, genes, regulatory regions, introns, exons, open-reading frames, binding sites, target nucleic acids and allele-specific nucleic acids.


A “probe,” as used herein, includes a nucleic acid fragment for examining a nucleic acid in a specimen using the hybridization reaction based on the complementarity of nucleic acid.


A “hybrid” as used herein, includes a double strand formed between any one of the abovementioned nucleic acid, within the same type, or across different types, including DNA-DNA, DNA-RNA, RNA-RNA or the like.


“Isolated” nucleic acids, as used herein, are separated from nucleic acids that normally flank the gene or nucleotide sequence (as in genomic sequences) and/or has been completely or partially purified from other transcribed sequences (e.g., as in an RNA library). For example, isolated nucleic acids of the disclosure can be substantially isolated with respect to the complex cellular milieu in which it naturally occurs, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized. In some instances, the isolated material can form part of a composition, for example, a crude extract containing other substances, buffer system or reagent mix. In some embodiments, the material can be purified to essential homogeneity using methods known in the art, for example, by polyacrylamide gel electrophoresis (PAGE) or column chromatography (e.g., HPLC). With regard to genomic DNA (gDNA), the term “isolated” also can refer to nucleic acids that are separated from the chromosome with which the genomic DNA is naturally associated. For example, the isolated nucleic acid molecule can contain less than about 250 kb, 200 kb, 150 kb, 100 kb, 75 kb, 50 kb, 25 kb, 10 kb, 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of the nucleotides that flank the nucleic acid molecule in the gDNA of the cell from which the nucleic acid molecule is derived.


Nucleic acids can be fused to other coding or regulatory sequences can be considered isolated. For example, recombinant DNA contained in a vector is included in the definition of “isolated” as used herein. In some embodiments, isolated nucleic acids can include recombinant DNA molecules in heterologous host cells or heterologous organisms, as well as partially or substantially purified DNA molecules in solution. Isolated nucleic acids also encompass in vivo and in vitro RNA transcripts of the DNA molecules of the present disclosure. An isolated nucleic acid molecule or nucleotide sequence can be synthesized chemically or by recombinant means. Such isolated nucleotide sequences can be useful, for example, in the manufacture of the encoded polypeptide, as probes for isolating homologous sequences (e.g., from other mammalian species), for gene mapping (e.g., by in situ hybridization with chromosomes), or for detecting expression of the gene, in tissue (e.g., human tissue), such as by Northern blot analysis or other hybridization techniques disclosed herein. The disclosure also pertains to nucleic acid sequences that hybridize under high stringency hybridization conditions, such as for selective hybridization, to a nucleotide sequence described herein Such nucleic acid sequences can be detected and/or isolated by allele- or sequence-specific hybridization (e.g., under high stringency conditions). Stringency conditions and methods for nucleic acid hybridizations are well known to the skilled person (see, e.g., Current Protocols in Molecular Biology, Ausubel, F. et al., John Wiley & Sons, (1998), and Kraus, M. and Aaronson, S., Methods Enzymol., 200:546-556 (1991), the entire teachings of which are incorporated by reference herein.


Calculations of “identity” or “percent identity” between two or more nucleotide or amino acid sequences can be determined by aligning the sequences for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first sequence). The nucleotides at corresponding positions are then compared, and the percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e. % identity=# of identical positions/total # of positions×100). For example, a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.


In some embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%, of the length of the reference sequence. The actual comparison of the two sequences can be accomplished by well-known methods, for example, using a mathematical algorithm. A non-limiting example of such a mathematical algorithm is described in Karlin, S. and Altschul, S., Proc. Natl. Acad. Sci. USA, 90-5873-5877 (1993). Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0), as described in Altschul, S. et al., Nucleic Acids Res., 25:3389-3402 (1997). When utilizing BLAST and Gapped BLAST programs, any relevant parameters of the respective programs (e.g., NBLAST) can be used. For example, parameters for sequence comparison can be set at score=100, word length=12, or can be varied (e.g., W=5 or W=20). Other examples include the algorithm of Myers and Miller, CABIOS (1989), ADVANCE, ADAM, BLAT, and FASTA. In some embodiments, the percent identity between two amino acid sequences can be accomplished using, for example, the GAP program in the GCG software package (Accelrys, Cambridge, UK).


“Probes” or “primers” can be oligonucleotides that hybridize in a base-specific manner to a complementary strand of a nucleic acid molecule. Probes can include primers, which can be a single-stranded oligonucleotide probe that can act as a point of initiation of template-directed DNA synthesis using methods including but not limited to, polymerase chain reaction (PCR) and ligase chain reaction (LCR) for amplification of a target sequence. Oligonucleotides, as described herein, can include segments or fragments of nucleic acid sequences, or their complements. In some embodiments, DNA segments can be between 5 and 10,000 contiguous bases, and can range from 5, 10, 12, 15, 20, or 25 nucleotides to 10, 15, 20, 25, 30, 40, 50, 100, 200, 500, 1000 or 10,000 nucleotides. In addition to DNA and RNA, probes and primers can include polypeptide nucleic acids (PNA), as described in Nielsen, P. et al., Science 254: 1497-1500 (1991). A probe or primer can comprise a region of nucleotide sequence that hybridizes to at least about 15, typically about 20-25, and in certain embodiments about 40, 50, 60 or 75, consecutive nucleotides of a nucleic acid molecule.


The present disclosure also provides isolated nucleic acids, for example, probes or primers, that contain a fragment or portion that can selectively hybridize to a nucleic acid that comprises, or consists of, a nucleotide sequence, wherein the nucleotide sequence can comprise at least one polymorphism or polymorphic allele contained in the genetic variations described herein or the wild-type nucleotide that is located at the same position, or the complements thereof. In some embodiments, the probe or primer can be at least 70% identical, at least 80% identical, at least 85% identical, at least 90% identical, or at least 95% identical, to the contiguous nucleotide sequence or to the complement of the contiguous nucleotide sequence.


In some embodiments, a nucleic acid probe can be an oligonucleotide capable of hybridizing with a complementary region of a gene associated with a condition (e.g., PML) containing a genetic variation described herein. The nucleic acid fragments of the disclosure can be used as probes or primers in assays such as those described herein.


The nucleic acids of the disclosure, such as those described above, can be identified and isolated using standard molecular biology techniques well known to the skilled person. In some embodiments, DNA can be amplified and/or can be labeled (e.g., radiolabeled, fluorescently labeled) and used as a probe for screening, for example, a cDNA library derived from an organism. cDNA can be derived from mRNA and can be contained in a suitable vector. For example, corresponding clones can be isolated, DNA obtained fallowing in vivo excision, and the cloned insert can be sequenced in either or both orientations by art-recognized methods to identify the correct reading frame encoding a polypeptide of the appropriate molecular weight. Using these or similar methods, the polypeptide and the DNA encoding the polypeptide can be isolated, sequenced and further characterized.


In some embodiments, nucleic acid can comprise one or more polymorphisms, variations, or mutations, for example, single nucleotide polymorphisms (SNPs), single nucleotide variations (SNVs), copy number variations (CNVs), for example, insertions, deletions, inversions, and translocations. In some embodiments, nucleic acids can comprise analogs, for example, phosphorothioates, phosphoramidates, methyl phosphonate, chiralmethyl phosphonates, 2-0-methyl ribonucleotides, or modified nucleic acids, for example, modified backbone residues or linkages, or nucleic acids combined with carbohydrates, lipids, polypeptide or other materials, or peptide nucleic acids (PNAs), for example, chromatin, ribosomes, and transcriptosomes. In some embodiments nucleic acids can comprise nucleic acids in various structures, for example, A DNA, B DNA, Z-form DNA, siRNA, tRNA, and ribozymes. In some embodiments, the nucleic acid may be naturally or non-naturally polymorphic, for example, having one or more sequence differences, for example, additions, deletions and/or substitutions, as compared to a reference sequence. In some embodiments, a reference sequence can be based on publicly available information, for example, the U.C. Santa Cruz Human Genome Browser Gateway (genome.ucsc.edu/cgi-bin/hgGateway) or the NCBI website (www.ncbi.nlm.nih.gov). In some embodiments, a reference sequence can be determined by a practitioner of the present disclosure using methods well known in the art, for example, by sequencing a reference nucleic acid.


In some embodiment a probe can hybridize to an allele, SNP, SNV, or CNV as described herein. In some embodiments, the probe can bind to another marker sequence associated with PML as described herein.


One of skill in the art would know how to design a probe so that sequence specific hybridization can occur only if a particular allele is present in a genomic sequence from a test nucleic acid sample. The disclosure can also be reduced to practice using any convenient genotyping method, including commercially available technologies and methods for genotyping particular genetic variations


Control probes can also be used, for example, a probe that binds a less variable sequence, for example, a repetitive DNA associated with a centromere of a chromosome, can be used as a control. In some embodiments, probes can be obtained from commercial sources. In some embodiments, probes can be synthesized, for example, chemically or in vitro, or made from chromosomal or genomic DNA through standard techniques. In some embodiments sources of DNA that can be used include genomic DNA, cloned DNA sequences, somatic cell hybrids that contain one, or a part of one, human chromosome along with the normal chromosome complement of the host, and chromosomes purified by flow cytometry or microdissection. The region of interest can be isolated through cloning, or by site-specific amplification using PCR.


One or more nucleic acids for example, a probe or primer, can also be labeled, for example, by direct labeling, to comprise a detectable label. A detectable label can comprise any label capable of detection by a physical, chemical, or a biological process for example, a radioactive label, such as 32P or 3H, a fluorescent label, such as FITC, a chromophore label, an affinity-ligand label, an enzyme label, such as alkaline phosphatase, horseradish peroxidase, or 12 galactosidase, an enzyme cofactor label, a hapten conjugate label, such as digoxigenin or dinitrophenyl, a Raman signal generating label, a magnetic label, a spin label, an epitope label, such as the FLAG or HA epitope, a luminescent label, a heavy atom label, a nanoparticle label, an electrochemical label, a light scattering label, a spherical shell label, semiconductor nanocrystal label, such as quantum dots (described in U.S. Pat. No. 6,207,392), and probes labeled with any other signal generating label known to those of skill in the art, wherein a label can allow the probe to be visualized with or without a secondary detection molecule. A nucleotide can be directly incorporated into a probe with standard techniques, for example, nick translation, random priming, and PCR labeling. A “signal,” as used herein, include a signal suitably detectable and measurable by appropriate means, including fluorescence, radioactivity, chemiluminescence, and the like.


Non-limiting examples of label moieties useful for detection include, without limitation, suitable enzymes such as horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; members of a binding pair that are capable of forming complexes such as streptavidin/biotin, avidin/biotin or an antigen/antibody complex including, for example, rabbit IgG and anti-rabbit IgG; fluorophores such as umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, tetramethyl rhodamine, eosin, green fluorescent protein, erythrosin, coumarin, methyl coumarin, pyrene, malachite green, stilbene, lucifer yellow, Cascade Blue, Texas Red, dichlorotriazinylamine fluorescein, dansyl chloride, phycoerythrin, fluorescent lanthanide complexes such as those including Europium and Terbium, cyanine dye family members, such as Cy3 and Cy5, molecular beacons and fluorescent derivatives thereof, as well as others known in the art as described, for example, in Principles of Fluorescence Spectroscopy, Joseph R. Lakowicz (Editor), Plenum Pub Corp, 2nd edition (July 1999) and the 6th Edition of the Molecular Probes Handbook by Richard P. Hoagland; a luminescent material such as luminol; light scattering or plasmon resonant materials such as gold or silver particles or quantum dots; or radioactive material include 14C, 123I, 124I, 125I, Tc99m, 32P, 33P, 35S or 3H.


Other labels can also be used in the methods of the present disclosure, for example, backbone labels. Backbone labels comprise nucleic acid stains that bind nucleic acids in a sequence independent manner. Non-limiting examples include intercalating dyes such as phenanthridines and acridines (e.g., ethidium bromide, propidium iodide, hexidium iodide, dihydroethidium, ethidium homodimer-1 and -2, ethidium monoazide, and ACMA); some minor grove binders such as indoles and imidazoles (e.g., Hoechst 33258, Hoechst 33342, Hoechst 34580 and DAPI); and miscellaneous nucleic acid stains such as acridine orange (also capable of intercalating), 7-AAD, actinomycin D, LDS751, and hydroxystilbamidine. All of the aforementioned nucleic acid stains are commercially available from suppliers such as Molecular Probes, Inc. Still other examples of nucleic acid stains include the following dyes from Molecular Probes: cyanine dyes such as SYTOX Blue, SYTOX Green, SYTOX Orange, POPO-1, POPO-3, YOYO-1, YOYO-3, TOTO-1, TOTO-3, JOJO-1, LOLO-1, BOBO-1, BOBO-3, PO-PRO-1, PO-PRO-3, BO-PRO-1, BO-PRO-3, TO-PRO-1, TO-PRO-3, TO-PRO-5, JO-PRO-1, LO-PRO-1, YO-PRO-1, YO-PRO-3, PicoGreen, OliGreen, RiboGreen, SYBR Gold, SYBR Green I, SYBR Green II, SYBR DX, SYTO-40, -41, -42, -43, -44, -45 (blue), SYTO-13, -16, -24, -21, -23, -12, -11, -20, -22, -15, -14, -25 (green), SYTO-81, -80, -82, -83, -84, -85 (orange), SYTO-64, -17, -59, -61, -62, -60, -63 (red).


In some embodiments, fluorophores of different colors can be chosen, for example, 7-amino-4-methylcoumarin-3-acetic acid (AMCA), 5-(and-6)-carboxy-X-rhodamine, lissamine rhodamine B, 5-(and-6)-carboxyfluorescein, fluorescein-5-isothiocyanate (FITC), 7-diethylaminocoumarin-3-carboxylic acid, tetramethylrhodamine-5-(and-6)-isothiocyanate, 5-(and-6)-carboxytetramethylrhodamine, 7-hydroxycoumarin-3-carboxylic acid, 6-[fluorescein 5-(and-6)-carboxamidolhexanoic acid, N-(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a diaza-3-indacenepropionic acid, eosin-5-isothiocyanate, erythrosin-5-isothiocyanate, TRITC, rhodamine, tetramethylrhodamine, R-phycoerythrin, Cy-3, Cy-5, Cy-7, Texas Red, Phar-Red, allophycocyanin (APC), and CASCADE™ blue acetylazide, such that each probe in or not in a set can be distinctly visualized. In some embodiments, fluorescently labeled probes can be viewed with a fluorescence microscope and an appropriate filter for each fluorophore, or by using dual or triple band-pass filter sets to observe multiple fluorophores. In some embodiments, techniques such as flow cytometry can be used to examine the hybridization pattern of the probes.


In other embodiments, the probes can be indirectly labeled, for example, with biotin or digoxygenin, or labeled with radioactive isotopes such as 32P and/or 3H. As a non-limiting example, a probe indirectly labeled with biotin can be detected by avidin conjugated to a detectable marker. For example, avidin can be conjugated to an enzymatic marker such as alkaline phosphatase or horseradish peroxidase. In some embodiments, enzymatic markers can be detected using colorimetric reactions using a substrate and/or a catalyst for the enzyme. In some embodiments, catalysts for alkaline phosphatase can be used, for example, 5-bromo-4-chloro-3-indolylphosphate and nitro blue tetrazolium. In some embodiments, a catalyst can be used for horseradish peroxidase, for example, diaminobenzoate.


One or more genes disclosed herein can be in conditions or molecular pathways related to various aspects of immune function including, but not limited to, Type I interferon response (e.g., PMID 26052098), B cell receptor pathway (e.g., Wikipathways WP23; PMID 22566564), RANKL/RANK signaling pathway (e.g., Wikipathways WP2018), TCR signaling pathway (e.g., Wikipathways WP69), NF-kB signaling (e.g., PMID 28362430), JAK-STAT pathway (e.g., PMID 28255960), post-translational modification biology such as ubiquitination via LUBAC (e.g., PMID 23104095, 24958845, 25086647, 26085218, 26111062, 26525107, 26848516, 26877205, 27178468, 27786304, 27892465), Aicardi-Goutieres syndrome (e.g., PMID 26052098), eosinophilia (e.g., PMID 27222657), congenital neutropenia (e.g., PMID 24753205), T cell receptor defects (e.g., PMID 25452106, 25636200, 26246585, 26379669, 26453379, 28400082), and autophagy defects (e.g., 19229298, 22984599, 23222957, 26917586, 26953272, 27588602). In some embodiments, one or more genes disclosed herein can be related to JC virus biology (e.g., PMID 15327898, 19282432, 19903823, 22984599, 25910481). In some embodiments, one or more genes disclosed herein can be antiviral immune response genes.









TABLE 27





Examplary pathways and biology for PML risk genes (96-gene panel)*




























Eosinophilia-







B cell

associated




Autoinflammatory
Autophagy
B cell
receptor
Deubiquitinase
immuno
JC virus


Genes
disease
defects
defects
pathway
pathway
deficiency
biology





AP3B1









APOL1






27042682


ASH1L









ATM


23765059


27222657
19903823,









25910481,









27042682


ATR






19903823,









25910481


BLM









CARD11


23765059
23765059,

27222657







WP23





CDKN1B









CHD7





27222657



CLCN7









DCLRE1C









DDX58









DOCK8


23765059


27222657



EGR1









EPG5

23222957,









26917586







ETF1









FPR2









GATA2


23765059






GFI1









HIVEP1









HIVEP2









HTR2A






27042682


IDO2









IFIH1









IFNGR2









IFNLR1









IGLL1


23765059
23765059





IKBKB
28469620

23765059
23765059,
26877205,








WP23
28362430




IL17F









IL1B




27892465

15327898


IL21R


23765059






IRAK4


23765059
23765059
28362430




ITSN2









JUN



WP23


27042682


KAT6B









KCTD7









LIG4





27222657



LRBA

26707784
23765059






MALL









MAPK3






27042682


MAVS









MCEE









MKL1









MYD88


23765059

28362430




NBN









NFKB1
28469620

23765059
WP23
26877205,









28362430




NOD2
28421071
26953272


28362430
27222657



NRIP1









PIAS1









PIAS2









PIK3CD









PIK3CD-AS1









PK3R1


23765059
WP23





PKHD1









PLCG2


23765059
WP23





PNPT1









POLA1









POLE


23765059






PRF1









PRKCB



WP23





PRKCD



WP23





PRKCH









PRKDC









PSTPIP1
28421071








PTEN









PTPRC



WP23





RABGEF1









RAD51






27042682


RAG1


23765059


27222657



RAG2


23765059


27222657



RIPK1
28469620



26877205,









27892465,









28362430




RIPK3




26877205,









27892465,









28362430




RNF168









RTEL1









SHARPIN
28469620

23765059

26877205,









27892465,









28362430




SKIV2L









SMAD4






27042682


STIM1


23765059






STIM2









STXBP2









TAP2









TBK1









TCIRG1









TICAM1




28362430




TLR3
28469620



28362430




TLR4
28469620



28362430




TNFRSF11A
28421071



28362430




TNFRSF13B


23765059






TNFRSF8









TP53









TRAF3









TRAFD1









TRPM2









VPS45









WEE1






27042682


ZAP70


23765059


27222657








RANKL/

T cell

Type I




PI3K
RANK
T cell
receptor
TLR
interferon


Genes
Osteopetrosis
signaling
pathway
defects
pathway
signaling
pathway





AP3B1
19782549,









24753205








APOL1









ASH1L









ATM






25692705


ATR






24799566


BLM









CARD11




WP69
25930993



CDKN1B









CHD7









CLCN7
23877423,









24753205








DCLRE1C









DDX58






25145756,









26052098,









26763980,









27821552


DOCK8



26379669





EGR1









EPG5









ETF1









FPR2









GATA2









GFI1
19782549,









24753205








HIVEP1









HIVEP2









HTR2A









IDO2









IFIH1






26052098,









27821552


IFNGR2









IFNLR1









IGLL1









IKBKB
21079651

WP2018

WP69
20404851,
17047224,








25930993
25145756


IL17F









IL1B




WP69
25930993
25145756,









26763980


IL21R









IRAK4





20404851,
25737587








25930993



ITSN2









JUN






25888367


KAT6B









KCTD7









LIG4









LRBA



26707784





MALL









MAPK3


WP2018

WP69
25930993



MAVS






19120474,









22626058,









22901541,









25145756,









26763980


MCEE









MKL1






26098208,









26098211


MYD88





20404851,
16474425,








25930993
18573338,









25145756


NBN









NFKB1
21079651
27616589
WP2018

WP69
20404851,
17047224,








25930993
25145756,









26763980


NOD2






26763980


NRIP1









PIAS1






24036127


PIAS2






21156324,









24036127


PIK3CD

27616589




20231019


PIK3CD-AS1

27616589







PK3R1

27616589
WP2018

WP69

26196376


PKHD1









PLCG2

27616589
WP2018
26379669
WP69
25930993



PNPT1









POLA1






27019227,









27821552


POLE









PRF1









PRKCB









PRKCD




WP69

11839738


PRKCH









PRKDC






23251783


PSTPIP1









PTEN

27616589







PTPRC

27616589




19673688,









25869642


RABGEF1









RAD51









RAG1

27616589







RAG2

27616589







RIPK1





20404851
25145756


RIPK3






25145756


RNF168









RTEL1









SHARPIN

25930993



20404851
22901541,









25145756


SKIV2L






25064072,









27821552


SMAD4









STIM1






22144678


STIM2









STXBP2









TAP2









TBK1





25930993
18573338,









22626058,









25145756,









26763980,









28049150


TCIRG1
23877423,









24753205








TICAM1





20404851,
19120474,








25930993
25145756,









28049150


TLR3





20404851,
19120474,








25930993
25145756,









28049150


TLR4





20404851,
25145756








25930993



TNFRSF11A
21079651,

WP2018


25930993
21527253,



23877423





25407789


TNFRSF13B





25930993



TNFRSF8









TP53









TRAF3
21079651

WP2018


25930993
22901541,









25723057,









26763980


TRAFD1
25992615




16221674
18849341


TRPM2









VPS45









WEE1









ZAP70

27616589


WP69





*PMID numbers are listed for curated PubMed references or Wikipathway ID number






Table 27 contains an exemplary pathways and biology for PML risk genes based on the 96-gene panel listed in Table 19. The genes disclosed herein, such as the genes in the 96-gene panel, can be grouped based on the pathway or biological processes they are involved in.


Methods of Screening


As used herein, screening a subject comprises diagnosing or determining, theranosing, or determining the susceptibility to developing (prognosing) a condition, for example, PML. In particular embodiments, the disclosure is a method of determining a presence of, or a susceptibility to, PML, by detecting at least one genetic variation in a sample from a subject as described herein. In some embodiments, detection of particular alleles, markers, variations, or haplotypes is indicative of a presence or susceptibility to a condition (e.g., PML).


While means for screening PML using a JCV antibody test exist, PML risk is not adequately assessed by the JCV antibody test alone. Thus there exists a need for an improved screening test for assessing the risk of developing PML. Described herein are methods of screening an individual for a risk of developing PML, including but not limited to, determining the identity and location of genetic variations, such as variations in nucleotide sequence and copy number, and the presence or absence of alleles or genotypes in one or more samples from one or more subjects using any of the methods described herein. In some embodiments, determining an association to having or developing PML can be performed by detecting particular variations that appear more frequently in test subjects compared to reference subjects and analyzing the molecular and physiological pathways these variations can affect.


Within any given population, there can be an absolute susceptibility of developing a disease or trait, defined as the chance of a person developing the specific disease or trait over a specified time-period. Susceptibility (e.g., being at-risk) is typically measured by looking at very large numbers of people, rather than at a particular individual. As described herein, certain copy number variations (genetic variations) and/or single nucleotide variations are found to be useful for susceptibility assessment of PML. Susceptibility assessment can involve detecting particular genetic variations in the genome of individuals undergoing assessment. Particular genetic variations are found more frequently in individuals with PML, than in individuals without PML. Therefore, these genetic variations have predictive value for detecting PML, or a susceptibility to PML, in an individual. Without intending to be limited by theory, it is believed that the genetic variations described herein to be associated with susceptibility of PML represent functional variants predisposing to the disease. In some embodiments, a genetic variation can confer a susceptibility of the condition, for example carriers of the genetic variation are at a different risk of the condition than non-carriers. In some embodiments, the presence of a genetic variation is indicative of increased susceptibility to PML.


In some embodiments, screening can be performed using any of the methods disclosed, alone or in combination. In some embodiments, screening can be performed using Polymerase Chain Reaction (PCR). In some embodiments screening can be performed using Array Comparative Genomic Hybridization (aCGH) to detect CNVs. In another preferred embodiment screening can be performed using exome sequencing to detect SNVs, indels, and in some cases CNVs using appropriate analysis algorithms. In another preferred embodiment screening is performed using high-throughput (also known as next generation) whole genome sequencing methods and appropriate algorithms to detect all or nearly all genetic variations present in a genomic DNA sample. In some embodiments, the genetic variation information as it relates to the current disclosure can be used in conjunction with any of the above mentioned symptomatic screening tests to screen a subject for PML, for example, using a combination of aCGH and/or sequencing with a JCV screening test, such as the JCV antibody test, CD62L test, or CSF IgM oligoclonal band test. In some embodiments, the L-selectin (CD62L) expressed by CD3+ CD4+ T cells in, for example, cryopreserved peripheral blood mononuclear cells (PBMCs), can be a biomarker for JCV screening. A CD62L expression can be correlated with the risk of PML.


In some embodiments, information from any of the above screening methods (e.g., specific symptoms, scoring matrix, or genetic variation data) can be used to define a subject as a test subject or reference subject. In some embodiments, information from any of the above screening methods can be used to associate a subject with a test or reference population, for example, a subject in a population.


In one embodiment, an association with PML can be determined by the statistical likelihood of the presence of a genetic variation in a subject with PML, for example, an unrelated individual or a first or second-degree relation of the subject. In some embodiments, an association with PML can be decided by determining the statistical likelihood of the absence of a genetic variation in an unaffected reference subject, for example, an unrelated individual or a first or second-degree relation of the subject. The methods described herein can include obtaining and analyzing a nucleic acid sample from one or more suitable reference subjects.


In the present context, the term screening comprises diagnosis, prognosis, and theranosis. Screening can refer to any available screening method, including those mentioned herein. As used herein, susceptibility can be proneness of a subject towards the development of PML, or towards being less able to resist PML than one or more control subjects. In some embodiments, susceptibility can encompass increased susceptibility. For example, particular nucleic acid variations of the disclosure as described herein can be characteristic of increased susceptibility to PML. In some embodiments, particular nucleic acid variations can confer decreased susceptibility, for example particular nucleic variations of the disclosure as described herein can be characteristic of decreased susceptibility to development of PML.


As described herein, a genetic variation predictive of susceptibility to or presence of PML can be one where the particular genetic variation is more frequently present in a group of subjects with the condition (affected), compared to the frequency of its presence in a reference group (control), such that the presence of the genetic variation is indicative of susceptibility to or presence of PML. In some embodiments, the reference group can be a population nucleic acid sample, for example, a random nucleic acid sample from the general population or a mixture of two or more nucleic acid samples from a population. In some embodiments, disease-free controls can be characterized by the absence of one or more specific disease-associated symptoms, for example, individuals who have not experienced symptoms associated with PML. In some embodiments, the disease-free control group is characterized by the absence of one or more disease-specific risk factors, for example, at least one genetic and/or environmental risk factor. In some embodiments, a reference sequence can be referred to for a particular site of genetic variation. In some embodiments, a reference allele can be a wild-type allele and can be chosen as either the first sequenced allele or as the allele from a control individual. In some embodiments, one or more reference subjects can be characteristically matched with one or more affected subjects, for example, with matched aged, gender or ethnicity.


A person skilled in the art can appreciate that for genetic variations with two or more alleles present in the population being studied, and wherein one allele can be found in increased frequency in a group of individuals with PML in the population, compared with controls, the other allele of the marker can be found in decreased frequency in the group of individuals with the trait or disease, compared with controls. In such a case, one allele of the marker, for example, the allele found in increased frequency in individuals with PML, can be the at-risk allele, while the other allele(s) can be a neutral or protective allele.


A genetic variant associated with PML can be used to predict the susceptibility of the disease for a given genotype. For any genetic variation, there can be one or more possible genotypes, for example, homozygote for the at-risk variant (e.g., in autosomal recessive disorders), heterozygote, and non-carrier of the at-risk variant. Autosomal recessive disorders can also result from two distinct genetic variants impacting the same gene such that the individual is a compound heterozygote (e.g., the maternal allele contains a different mutation than the paternal allele). Compound heterozygosity may result from two different SNVs, two different CNVs, an SNV and a CNV, or any combination of two different genetic variants but each present on a different allele for the gene. For X-linked genes, males who possess one copy of a variant-containing gene may be affected, while carrier females, who also possess a wild-type gene, may remain unaffected. In some embodiments, susceptibility associated with variants at multiple loci can be used to estimate overall susceptibility. For multiple genetic variants, there can be k (k=3{circumflex over ( )}n*2{circumflex over ( )}P) possible genotypes; wherein n can be the number of autosomal loci and p can be the number of gonosomal (sex chromosomal) loci. Overall susceptibility assessment calculations can assume that the relative susceptibilities of different genetic variants multiply, for example, the overall susceptibility associated with a particular genotype combination can be the product of the susceptibility values for the genotype at each locus. If the susceptibility presented is the relative susceptibility for a person, or a specific genotype for a person, compared to a reference population, then the combined susceptibility can be the product of the locus specific susceptibility values and can correspond to an overall susceptibility estimate compared with a population. If the susceptibility for a person is based on a comparison to non-carriers of the at-risk allele, then the combined susceptibility can correspond to an estimate that compares the person with a given combination of genotypes at all loci to a group of individuals who do not carry at-risk variants at any of those loci. The group of non-carriers of any at-risk variant can have the lowest estimated susceptibility and can have a combined susceptibility, compared with itself, for example, non-carriers, of 1.0, but can have an overall susceptibility, compared with the population, of less than 1.0.


Overall risk for multiple risk variants can be performed using standard methodology. Genetic variations described herein can form the basis of risk analysis that combines other genetic variations known to increase risk of PML, or other genetic risk variants for PML. In certain embodiments of the disclosure, a plurality of variants (genetic variations, variant alleles, and/or haplotypes) can be used for overall risk assessment. These variants are in some embodiments selected from the genetic variations as disclosed herein. Other embodiments include the use of the variants of the present disclosure in combination with other variants known to be useful for screening a susceptibility to PML. In such embodiments, the genotype status of a plurality of genetic variations, markers and/or haplotypes is determined in an individual, and the status of the individual compared with the population frequency of the associated variants, or the frequency of the variants in clinically healthy subjects, such as age-matched and sex-matched subjects.


Methods such as the use of available algorithms and software can be used to identify, or call, significant genetic variations, including but not limited to, algorithms of DNA Analytics or DNAcopy, iPattern and/or QuantiSNP. In some embodiments, a threshold logratio value can be used to determine losses and gains. For example, using DNA Analytics, a log2 ratio cutoff of ≥0.5 and ≥0.5 to classify CNV gains and losses respectively can be used. For example, using DNA Analytics, a log2 ratio cutoff of ≥0.25 and ≥0.25 to classify CNV gains and losses respectively can be used. As a further example, using DNAcopy, a log2 ratio cutoff of ≥0.35 and ≥0.35 to classify CNV gains and losses respectively can be used. For example, an Aberration Detection Module 2 (ADM2) algorithm, such as that of DNA Analytics 4.0.85 can be used to identify, or call, significant genetic variations. In some embodiments, two or more algorithms can be used to identify, or call, significant genetic variations. For example, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more algorithms can be used to identify, or call, significant genetic variations. In another embodiment, the log 2 ratio of one or more individual probes on a microarray can be used to identify significant genetic variations, such as the presence of homozygously deleted regions in a subject's genome. In some embodiments, significant genetic variations can be CNVs.


CNVs detected by two or more algorithms can be defined as stringent and can be utilized for further analyses. In some embodiments, the information and calls from two or more of the methods described herein can be compared to each other to identify significant genetic variations more or less stringently. For example, CNV calls generated by two or more of DNA Analytics, Aberration Detection Module 2 (ADM2) algorithms, and DNAcopy algorithms can be defined as stringent CNVs. In some embodiments significant or stringent genetic variations can be tagged as identified or called if it can be found to have a minimal reciprocal overlap to a genetic variation detected by one or more platforms and/or methods described herein. For example, a minimum of 50% reciprocal overlap can be used to tag the CNVs as identified or called. For example, significant or stringent genetic variations can be tagged as identified or called if it can be found to have a reciprocal overlap of more than about 50%, 55% 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%, 99%, or equal to 100%, to a genetic variation detected by one or more platforms and/or methods described herein. For example, significant or stringent genetic variations can be tagged as identified or called if it can be found to have a reciprocal overlap of more than about 50% reciprocal overlap to a genetic variation detected by one or more platforms and/or methods described herein. In another embodiment, genetic variations can be detected from the log 2 ratio values calculated for individual probes present on an aCGH microarray via a statistical comparison of the probe's log 2 ratio value in a cohort of subjects with PML to the probe's log 2 ratio value in a cohort of subjects without PML.


In some embodiments, a threshold log ratio value can be used to determine losses and gains. A log ratio value can be any log ratio value; for example, a log ratio value can be a log 2 ratio or a log 10 ratio. In some embodiments, a CNV segment whose median log 2 ratio is less than or equal to a log 2 ratio threshold value can be classified as a loss. For example, any segment whose median log 2 ratio is less than or equal to −0.1, −0.11, −0.12, −0.13, −0.14, −0.15, −0.16, −0.17, −0.18, −0.19, −0.2, −0.21, −0.22, −0.23, −0.24, −0.25, −0.26, −0.27, −0.28, −0.29, −0.3, −0.31, −0.32, −0.33, −0.34, −0.35, −0.36, −0.37, −0.38, −0.39, −0.4, −0.41, −0.42, −0.43, −0.44, −0.45, −0.46, −0.47, −0.48, −0.49, −0.5, −0.55, −0.6, −0.65, −0.7, −0.75, −0.8, −0.85, −0.9, −0.95, −1, −1.1, −1.2, −1.3, −1.4, −1.5, −1.6, −1.7, −1.8, −1.9, −2, −2.1, −2.2, −2.3, −2.4, −2.5, −2.6, −2.7, −2.8, −2.9, −3, −3.1, −3.2, −3.3, −3.4, −3.5, −3.6, −3.7, −3.8, −3.9, −4, −4.1, −4.2, −4.3, −4.4, −4.5, −4.6, −4.7, −4.8, −4.9, −5, −5.5, −6, −6.5, −7, −7.5, −8, −8.5, −9, −9.5, −10, −11, −12, −13, −14, −15, −16, −17, −18, −19, −20 or less, can be classified as a loss.


In some embodiments, one algorithm can be used to call or identify significant genetic variations, wherein any segment whose median log 2 ratio was less than or equal to −0.1, −0.11, −0.12, −0.13, −0.14, −0.15, −0.16, −0.17, −0.18, −0.19, −0.2, −0.21, −0.22, −0.23, −0.24, −0.25, −0.26, −0.27, −0.28, −0.29, −0.3, −0.31, −0.32, −0.33, −0.34, −0.35, −0.36, −0.37, −0.38, −0.39, −0.4, −0.41, −0.42, −0.43, −0.44, −0.45, −0.46, −0.47, −0.48, −0.49, −0.5, −0.55, −0.6, −0.65, −0.7, −0.75, −0.8, −0.85, −0.9, −0.95, −1, −1.1, −1.2, −1.3, −1.4, −1.5, −1.6, −1.7, −1.8, −1.9, −2, −2.1, −2.2, −2.3, −2.4, −2.5, −2.6, −2.7, −2.8, −2.9, −3, −3.1, −3.2, −3.3, −3.4, −3.5, −3.6, −3.7, −3.8, −3.9, −4, −4.1, −4.2, −4.3, −4.4, −4.5, −4.6, −4.7, −4.8, −4.9, −5, −5.5, −6, −6.5, −7, −7.5, −8, −8.5, −9, −9.5, −10, −11, −12, −13, −14, −15, −16, −17, −18, −19, −20 or less, can be classified as a loss. For example, any CNV segment whose median log 2 ratio is less than −0.35 as determined by DNAcopy can be classified as a loss. For example, losses can be determined according to a threshold log 2 ratio, which can be set at −0.35. In another embodiment, losses can be determined according to a threshold log 2 ratio, which can be set at −0.5.


In some embodiments, two algorithms can be used to call or identify significant genetic variations, wherein any segment whose median log 2 ratio is less than or equal to −0.1, −0.11, −0.12, −0.13, −0.14, −0.15, −0.16, −0.17, −0.18, −0.19, −0.2, −0.21, −0.22, −0.23, −0.24, −0.25, −0.26, −0.27, −0.28, −0.29, −0.3, −0.31, −0.32, −0.33, −0.34, −0.35, −0.36, −0.37, −0.38, −0.39, −0.4, −0.41, −0.42, −0.43, −0.44, −0.45, −0.46, −0.47, −0.48, −0.49, −0.5, −0.55, −0.6, −0.65, −0.7, −0.75, −0.8, −0.85, −0.9, −0.95, −1, −1.1, −1.2, −1.3, −1.4, −1.5, −1.6, −1.7, −1.8, −1.9, −2, −2.1, −2.2, −2.3, −2.4, −2.5, −2.6, −2.7, −2.8, −2.9, −3, −3.1, −3.2, −3.3, −3.4, −3.5, −3.6, −3.7, −3.8, −3.9, −4, −4.1, −4.2, −4.3, −4.4, −4.5, −4.6, −4.7, −4.8, −4.9, −5, −5.5, −6, −6.5, −7, −7.5, −8, −8.5, −9, −9.5, −10, −11, −12, −13, −14, −15, −16, −17, −18, −19, −20 or less, as determined by one algorithm, and wherein any segment whose median log 2 ratio is less than or equal to −0.1, −0.11, −0.12, −0.13, −0.14, −0.15, −0.16, −0.17, −0.18, −0.19, −0.2, −0.21, −0.22, −0.23, −0.24, −0.25, −0.26, −0.27, −0.28, −0.29, −0.3, −0.31, −0.32, −0.33, −0.34, −0.35, −0.36, −0.37, −0.38, −0.39, −0.4, −0.41, −0.42, −0.43, −0.44, −0.45, −0.46, −0.47, −0.48, −0.49, −0.5, −0.55, −0.6, −0.65, −0.7, −0.75, −0.8, −0.85, −0.9, −0.95, −1, −1.1, −1.2, −1.3, −1.4, −1.5, −1.6, −1.7, −1.8, −1.9, −2, −2.1, −2.2, −2.3, −2.4, −2.5, −2.6, −2.7, −2.8, −2.9, −3, −3.1, −3.2, −3.3, −3.4, −3.5, −3.6, −3.7, −3.8, −3.9, −4, −4.1, −4.2, −4.3, −4.4, −4.5, −4.6, −4.7, −4.8, −4.9, −5, −5.5, −6, −6.5, −7, −7.5, −8, −8.5, −9, −9.5, −10, −11, −12, −13, −14, −15, −16, −17, −18, −19, −20, or less, as determined by the other algorithm can be classified as a loss. For example, CNV calling can comprise using the Aberration Detection Module 2 (ADM2) algorithm and the DNAcopy algorithm, wherein losses can be determined according to a two threshold log 2 ratios, wherein the Aberration Detection Module 2 (ADM2) algorithm log 2 ratio can be −0.25 and the DNAcopy algorithm log 2 ratio can be −0.41.


In some embodiments, the use of two algorithms to call or identify significant genetic variations can be a stringent method. In some embodiments, the use of two algorithms to call or identify significant genetic variations can be a more stringent method compared to the use of one algorithm to call or identify significant genetic variations.


In some embodiments, any CNV segment whose median log 2 ratio is greater than a log 2 ratio threshold value can be classified as a gain. For example, any segment whose median log 2 ratio is greater than 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.55, 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, or more can be classified as a gain.


In some embodiments, one algorithm can be used to call or identify significant genetic variations, wherein any segment whose median log 2 ratio is greater than or equal to 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.55, 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, or more can be classified as a gain. For example, any CNV segment whose median log 2 ratio is greater than 0.35 as determined by DNAcopy can be classified as a gain. For example, gains can be determined according to a threshold log 2 ratio, which can be set at 0.35. In another embodiment, gains can be determined according to a threshold log 2 ratio, which can be set at 0.5.


In some embodiments, two algorithms can be used to call or identify significant genetic variations, wherein any segment whose median log 2 ratio is greater than or equal to 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, 0.5, 0.55, 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3 or more, as determined by one algorithm, and wherein any segment whose median log 2 ratio is greater than or equal to 0.1, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.21, 0.22, 0.23, 0.24, 0.25, 0.26, 0.27, 0.28, 0.29, 0.3, 0.31, 0.32, 0.33, 0.34, 0.35, 0.36, 0.37, 0.38, 0.39, 0.4, 0.41, 0.42, 0.43, 0.44, 0.45, 0.46, 0.47, 0.48, 0.49, or 0.5, 0.55, 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3, or more, as determined by the other algorithm the can be classified as a gain. For example, CNV calling can comprise using the Aberration Detection Module 2 (ADM2) algorithm and the DNAcopy algorithm, wherein gains can be determined according to a two threshold log 2 ratios, wherein the Aberration Detection Module 2 (ADM2) algorithm log 2 ratio can be 0.25 and the DNAcopy algorithm log 2 ratio can be 0.32.


Any CNV segment whose absolute (median log-ratio/mad) value is less than 2 can be excluded (not identified as a significant genetic variation). For example, any CNV segment whose absolute (median log-ratio/mad) value is less than 2, 1.9, 1.8, 1.7, 1.6, 1.5, 1.4, 1.3, 1.2, 1.1, 1, 0.9, 0.8, 0.7, 0.6, or 0.5 or less can be excluded.


In some embodiments, multivariate analyses or joint risk analyses, including the use of multiplicative model for overall risk assessment, can subsequently be used to determine the overall risk conferred based on the genotype status at the multiple loci. Use of a multiplicative model, for example, assuming that the risk of individual risk variants multiply to establish the overall effect, allows for a straight-forward calculation of the overall risk for multiple markers. The multiplicative model is a parsimonious model that usually fits the data of complex traits reasonably well. Deviations from multiplicity have been rarely described in the context of common variants for common diseases, and if reported are usually only suggestive since very large sample sizes can be required to be able to demonstrate statistical interactions between loci. Assessment of risk based on such analysis can subsequently be used in the methods, uses and kits of the disclosure, as described herein.


In some embodiments, the significance of increased or decreased susceptibility can be measured by a percentage. In some embodiments, a significant increased susceptibility can be measured as a relative susceptibility of at least 1.2, including but not limited to: at least 1.3, at least 1.4, at least 1.5, at least 1.6, at least 1.7, at least 1.8, at least 1.9, at least 2.0, at least 2.5, at least 3.0, at least 4.0, at least 5.0, at least 6.0, at least 7.0, at least 8.0, at least 9.0, at least 10.0, and at least 15.0. In some embodiments, a relative susceptibility of at least 2.0, at least 3.0, at least 4.0, at least, 5.0, at least 6.0, or at least 10.0 is significant. Other values for significant susceptibility are also contemplated, for example, at least 2.5, 3.5, 4.5, 5.5, or any suitable other numerical values, wherein the values are also within scope of the present disclosure. In some embodiments, a significant increase in susceptibility is at least about 20%, including but not limited to about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 150%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000%, and 1500%. In one particular embodiment, a significant increase in susceptibility is at least 100%. In other embodiments, a significant increase in susceptibility is at least 200%, at least 300%, at least 400%, at least 500%, at least 700%, at least 800%, at least 900% and at least 1000%. Other cutoffs or ranges as deemed suitable by the person skilled in the art to characterize the disclosure are also contemplated, and those are also within scope of the present disclosure. In certain embodiments, a significant increase in susceptibility is characterized by a p-value, such as a p-value of less than 0.5, less than 0.4, less than 0.3, less than 0.2, less than 0.1, less than 0.05, less than 0.01, less than 0.001, less than 0.0001, less than 0.00001, less than 0.000001, less than 0.0000001, less than 0.00000001, or less than 0.000000001.


In some embodiments, an individual who is at a decreased susceptibility for or the lack of presence of a condition (e.g., PML) can be an individual in whom at least one genetic variation, conferring decreased susceptibility for or the lack of presence of the condition is identified. In some embodiments, the genetic variations conferring decreased susceptibility are also protective. In one aspect, the genetic variations can confer a significant decreased susceptibility of or lack of presence of PML.


In some embodiments, significant decreased susceptibility can be measured as a relative susceptibility of less than 0.9, including but not limited to less than 0.9, less than 0.8, less than 0.7, less than 0.6, less than 0.5, less than 0.4, less than 0.3, less than 0.2 and less than 0.1. In some embodiments, the decrease in susceptibility is at least 20%, including but not limited to at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% and at least 98%. Other cutoffs or ranges as deemed suitable by the person, skilled in the art to characterize the disclosure are however also contemplated, and those are also within scope of the present disclosure. In certain embodiments, a significant decrease in susceptibility is characterized by a p-value, such as a p-value of less than 0.05, less than 0.01, less than 0.001, less than 0.0001, less than 0.00001, less than 0.000001, less than 0.0000001, less than 0.00000001, or less than 0.000000001. Other tests for significance can be used, for example, a Fisher-exact test. Other statistical tests of significance known to the skilled person are also contemplated and are also within scope of the disclosure.


In some preferred embodiments, the significance of increased or decreased susceptibility can be determined according to the ratio of measurements from a test subject to a reference subject. In some embodiments, losses or gains of one or more CNVs can be determined according to a threshold log2 ratio determined by these measurements. In some embodiments, a log2 ratio value greater than 0.35, or 0.5, is indicative of a gain of one or more CNVs. In some embodiments, a log2 ratio value less than −0.35, or −0.5, is indicative of a loss of one or more CNVs. In some embodiments, the ratio of measurements from a test subject to a reference subject may be inverted such that the log 2 ratios of copy number gains are negative and the log 2 ratios of copy number losses are positive.


In some embodiments, the combined or overall susceptibility associated with a plurality of variants associated with PML can also be assessed; for example, the genetic variations described herein to be associated with susceptibility to PML can be combined with other common genetic risk factors. Combined risk for such genetic variants can be estimated in an analogous fashion to the methods described herein.


Calculating risk conferred by a particular genotype for the individual can be based on comparing the genotype of the individual to previously determined risk expressed, for example, as a relative risk (RR) or an odds ratio (OR), for the genotype, for example, for a heterozygous carrier of an at-risk variant for PML. An odds ratio can be a statistical measure used as a metric of causality. For example, in genetic disease research it can be used to convey the significance of a variant in a disease cohort relative to an unaffected/normal cohort. The calculated risk for the individual can be the relative risk for a subject, or for a specific genotype of a subject, compared to the average population. The average population risk can be expressed as a weighted average of the risks of different genotypes, using results from a reference population, and the appropriate calculations to calculate the risk of a genotype group relative to the population can then be performed. Alternatively, the risk for an individual can be based on a comparison of particular genotypes, for example, heterozygous and/or homozygous carriers of an at-risk allele of a marker compared with non-carriers of the at-risk allele (or pair of alleles in the instance of compound heterozygous variants, wherein one variant impacts the maternally inherited allele and the other impacts the paternally inherited allele). Using the population average can, in certain embodiments, be more convenient, since it provides a measure that can be easy to interpret for the user, for example, a measure that gives the risk for the individual, based on his/her genotype, compared with the average in the population.


In some embodiments, the OR value can be calculated as follows: OR=(A/(N1−A))/(U/(N2−U)), where A=number of affected cases with variant, N1=total number of affected cases, U=number of unaffected cases with variant and N2=total number of unaffected cases. In circumstances where U=0, it is conventional to set U=1, so as to avoid infinities. In some preferred embodiments, the OR can be calculated essentially as above, except that where U or A=0, 0.5 is added to all of A, N1, U, N2. In another embodiment, a Fisher's Exact Test (FET) can be calculated using standard methods. In another embodiment, the p-values can be corrected for false discovery rate (FDR) using the Benjamini-Hochberg method (Benjamini Y. and Hochberg Y., J. Royal Statistical Society 57:289 (1995); Osborne J. A. and Barker C. A. (2007)).


In certain embodiments of the disclosure, a genetic variation is correlated to PML by referencing genetic variation data to a look-up table that comprises correlations between the genetic variation and PML. The genetic variation in certain embodiments comprises at least one indication of the genetic variation. In some embodiments, the table comprises a correlation for one genetic variation. In other embodiments, the table comprises a correlation for a plurality of genetic variations in both scenarios, by referencing to a look-up table that gives an indication of a correlation between a genetic variation and PML, a risk for PML, or a susceptibility to PML, can be identified in the individual from whom the nucleic acid sample is derived.


The present disclosure also pertains to methods of clinical screening, for example, diagnosis, prognosis, or theranosis of a subject performed by a medical professional using the methods disclosed herein. In other embodiments, the disclosure pertains to methods of screening performed by a layman. The layman can be a customer of a genotyping, microarray, exome sequencing, or whole genome sequencing service provider. The layman can also be a genotype, microarray, exome sequencing, or whole genome sequencing service provider, who performs genetic analysis on a DNA sample from an individual, in order to provide service related to genetic risk factors for particular traits or diseases, based on the genotype status of the subject obtained from use of the methods described herein. The resulting genotype or genetic information can be made available to the individual and can be compared to information about PML or risk of developing PML associated with one or various genetic variations, including but not limited to, information from public or private genetic variation databases or literature and scientific publications. The screening applications of PML-associated genetic variations, as described herein, can, for example, be performed by an individual, a health professional, or a third party, for example a service provider who interprets genotype information from the subject. In some embodiments the genetic analysis is performed in a CLIA-certified laboratory (i.e. the federal regulatory standards the U.S. that are specified in the Clinical Laboratory Improvement Amendments, administered by the Centers for Medicare and Medicaid Services) or equivalent laboratories in Europe and elsewhere in the world.


The information derived from analyzing sequence data can be communicated to any particular body, including the individual from which the nucleic acid sample or sequence data is derived, a guardian or representative of the individual, clinician, research professional, medical professional, service provider, and medical insurer or insurance company. Medical professionals can be, for example, doctors, nurses, medical laboratory technologists, and pharmacists. Research professionals can be, for example, principle investigators, research technicians, postdoctoral trainees, and graduate students.


In some embodiments, a professional can be assisted by determining whether specific genetic variants are present in a nucleic acid sample from a subject, and communicating information about genetic variants to a professional. After information about specific genetic variants is reported, a medical professional can take one or more actions that can affect subject care. For example, a medical professional can record information in the subject's medical record (e.g., electronic health record or electronic medical record, including, but not limited to, country-scale health services such as the National Health Service in the United Kingdom) regarding the subject's risk of developing PML. In some embodiments, a medical professional can record information regarding risk assessment, or otherwise transform the subject's medical record, to reflect the subject's current medical condition. In some embodiments, a medical professional can review and evaluate a subject's entire medical record and assess multiple treatment strategies for clinical intervention of a subject's condition. In another embodiment, information can be recorded in the context of the system developed by the World Health Organization (WHO), the International Statistical Classification of Diseases and Related Health Problems (ICD), which is currently using the 10th revision (ICD-10 codes). For example, the ICD-10 code for PML is A81.2, whereas the ICD-10 code for multiple sclerosis is G35.


A medical professional can initiate or modify treatment after receiving information regarding a subject's screening for PML, for example. In some embodiments, a medical professional can recommend a change in therapy or exclude a therapy. In some embodiments, a medical professional can enroll a subject in a clinical trial for, by way of example, detecting correlations between a haplotype as described herein and any measurable or quantifiable parameter relating to the outcome of the treatment as described above.


In some embodiments, a medical professional can communicate information regarding a subject's screening of developing PML to a subject or a subject's family. In some embodiments, a medical professional can provide a subject and/or a subject's family with information regarding PML and risk assessment information, including treatment options, and referrals to specialists. In some embodiments, a medical professional can provide a copy of a subject's medical records to a specialist. In some embodiments, a research professional can apply information regarding a subject's risk of developing PML to advance scientific research. In some embodiments, a research professional can obtain a subject's haplotype as described herein to evaluate a subject's enrollment, or continued participation, in a research study or clinical trial. In some embodiments, a research professional can communicate information regarding a subject's screening of PML to a medical professional. In some embodiments, a research professional can refer a subject to a medical professional.


Any appropriate method can be used to communicate information to another person. For example, information can be given directly or indirectly to a professional and a laboratory technician can input a subject's genetic variation as described herein into a computer-based record. In some embodiments, information is communicated by making a physical alteration to medical or research records. For example, a medical professional can make a permanent notation or flag a medical record for communicating the risk assessment to other medical professionals reviewing the record. In addition, any type of communication can be used to communicate the risk assessment information. For example, mail, e-mail, telephone, and face-to-face interactions can be used. The information also can be communicated to a professional by making that information electronically available to the professional. For example, the information can be communicated to a professional by placing the information on a computer database such that the professional can access the information. In addition, the information can be communicated to a hospital, clinic, or research facility serving as an agent for the professional.


Results of these tests, and optionally interpretive information, can be returned to the subject, the health care provider or to a third party. The results can be communicated to the tested subject, for example, with a prognosis and optionally interpretive materials that can help the subject understand the test results and prognosis; used by a health care provider, for example, to determine whether to administer a specific drug, or whether a subject should be assigned to a specific category, for example, a category associated with a specific disease endophenotype, or with drug response or non-response; used by a third party such as a healthcare payer, for example, an insurance company or HMO, or other agency, to determine whether or not to reimburse a health care provider for services to the subject, or whether to approve the provision of services to the subject. For example, the healthcare payer can decide to reimburse a health care provider for treatments for PML if the subject has PML or has an increased risk of developing PML.


Also provided herein are databases that include a list of genetic variations as described herein, and wherein the list can be largely or entirely limited to genetic variations identified as useful for screening PML as described herein. The list can be stored, for example, on a flat file or computer-readable medium. The databases can further include information regarding one or more subjects, for example, whether a subject is affected or unaffected, clinical information such as endophenotype, age of onset of symptoms, any treatments administered and outcomes, for example, data relevant to pharmacogenomics, diagnostics, prognostics or theranostics, and other details, for example, data about the disorder in the subject, or environmental (e.g., including, but not limited to, infection or a history of infection with HIV or JCV) or other genetic factors. The databases can be used to detect correlations between a particular haplotype and the information regarding the subject.


The methods described herein can also include the generation of reports for use, for example, by a subject, care giver, or researcher, that include information regarding a subject's genetic variations, and optionally further information such as treatments administered, treatment history, medical history, predicted response, and actual response. The reports can be recorded in a tangible medium, e.g., a computer-readable disk, a solid state memory device, or an optical storage device.


Methods of Screening Using Variations in RNA and/or Polypeptides


In some embodiments of the disclosure, screening of PML can be made by examining or comparing changes in expression, localization, binding partners, and composition of a polypeptide encoded by a nucleic acid variant associated with PML, for example, in those instances where the genetic variations of the present disclosure results in a change in the composition or expression of the polypeptide and/or RNA, for example, mRNAs, microRNAs (miRNAs), and other noncoding RNAs (ncRNAs). Thus, screening of PML can be made by examining expression and/or composition of one of these polypeptides and/or RNA, or another polypeptide and/or RNA encoded by a nucleic acid associated with PML, in those instances where the genetic variation of the present disclosure results in a change in the expression, localization, binding partners, and/or composition of the polypeptide and/or RNA. In some embodiments, screening can comprise diagnosing a subject. In some embodiments, screening can comprise determining a prognosis of a subject, for example determining the susceptibility of developing PML. In some embodiments, screening can comprise theranosing a subject.


The genetic variations described herein that show association to PML can play a role through their effect on one or more of these genes, either by directly impacting one or more genes or influencing the expression of one or more nearby genes. For example, while not intending to be limited by theory, it is generally expected that a deletion of a chromosomal segment comprising a particular gene, or a fragment of a gene, can either result in an altered composition or expression, or both, of the encoded polypeptide and/or mRNA. Likewise, duplications, or high number copy number variations, are in general expected to result in increased expression of encoded polypeptide and/or RNA if the gene they are expressed from is fully encompassed within the duplicated (or triplicated, or even higher copy number gains) genomic segment, or conversely can result in decreased expression or a disrupted RNA or polypeptide if one or both breakpoints of the copy number gain disrupt a given gene. Other possible mechanisms affecting genes within a genetic variation region include, for example, effects on transcription, effects on RNA splicing, alterations in relative amounts of alternative splice forms of mRNA, effects on RNA stability, effects on transport from the nucleus to cytoplasm, and effects on the efficiency and accuracy of translation. Thus, DNA variations can be detected directly, using the subjects unamplified or amplified genomic DNA, or indirectly, using RNA or DNA obtained from the subject's tissue(s) that are present in an aberrant form or expression level as a result of the genetic variations of the disclosure showing association to PML. In another embodiment, DNA variations can be detected indirectly using a polypeptide or protein obtained from the subject's tissue(s) that is present in an aberrant form or expression level as a result of genetic variations of the disclosure showing association to the PML. In another embodiment, an aberrant form or expression level of a polypeptide or protein that results from one or more genetic variations of the disclosure showing association to PML can be detected indirectly via another polypeptide or protein present in the same biological/cellular pathway that is modulated or interacts with said polypeptide or protein that results from one or more genetic variations of the disclosure. In some embodiments, the genetic variations of the disclosure showing association to PML can affect the expression of a gene within the genetic variation region. In some embodiments, a genetic variation affecting an exonic region of a gene can affect, disrupt, or modulate the expression of the gene. In some embodiments, a genetic variation affecting an intronic or intergenic region of a gene can affect, disrupt, or modulate the expression of the gene.


Certain genetic variation regions can have flanking duplicated segments, and genes within such segments can have altered expression and/or composition as a result of such genomic alterations. Regulatory elements affecting gene expression can be located far away, even as far as tens or hundreds of kilobases away, from the gene that is regulated by said regulatory elements. Thus, in some embodiments, regulatory elements for genes that are located outside the gene (e.g., upstream or downstream of the gene) can be located within the genetic variation, and thus be affected by the genetic variation. It is thus contemplated that the detection of the genetic variations described herein, can be used for assessing expression for one or more of associated genes not directly impacted by the genetic variations. In some embodiments, a genetic variation affecting an intergenic region of a gene can affect, disrupt, or modulate the expression of a gene located elsewhere in the genome, such as described above. For example, a genetic variation affecting an intergenic region of a gene can affect, disrupt, or modulate the expression of a transcription factor, located elsewhere in the genome, which regulates the gene. Regulatory elements can also be located within a gene, such as within intronic regions, and similarly impact the expression level of the gene and ultimately the protein expression level without changing the structure of the protein. The effects of genetic variants on regulatory elements can manifest in a tissue-specific manner; for example, one or more transcription factors that bind to the regulatory element that is impacted by one or more genetic variations may be expressed at higher concentration in neurons as compared to skin cells (i.e., the impact of the one or more genetic variations may be primarily evident in neuronal cells).


In some embodiments, genetic variations of the disclosure showing association to PML can affect protein expression at the translational level. It can be appreciated by those skilled in the art that this can occur by increased or decreased expression of one or more microRNAs (miRNAs) that regulates expression of a protein known to be important, or implicated, in the cause, onset, or progression of PML. Increased or decreased expression of the one or more miRNAs can result from gain or loss of the whole miRNA gene, disruption or impairment of a portion of the gene (e.g., by an indel or CNV), or even a single base change (SNP or SNV) that produces an altered, non-functional or aberrant functioning miRNA sequence. It can also be appreciated by those skilled in the art that the expression of protein, for example, one known to cause PML by increased or decreased expression, can result due to a genetic variation that results in alteration of an existing miRNA binding site within the polypeptide's mRNA transcript, or even creates a new miRNA binding site that leads to aberrant polypeptide expression.


A variety of methods can be used for detecting polypeptide composition and/or expression levels, including but not limited to enzyme linked immunosorbent assays (ELISA), Western blots, spectroscopy, mass spectrometry, peptide arrays, colorimetry, electrophoresis, isoelectric focusing, immunoprecipitations, immunoassays, and immunofluorescence and other methods well-known in the art. A test nucleic acid sample from a subject can be assessed for the presence of an alteration in the expression and/or an alteration in composition of the polypeptide encoded by a nucleic acid associated with PML. An “alteration” in the polypeptide expression or composition, as used herein, refers to an alteration in expression or composition in a test nucleic acid sample, as compared to the expression or composition of the polypeptide in a control nucleic acid sample. Such alteration can, for example, be an alteration in the quantitative polypeptide expression or can be an alteration in the qualitative polypeptide expression, for example, expression of a mutant polypeptide or of a different splicing variant, or a combination thereof. In some embodiments, screening of PML can be made by detecting a particular splicing variant encoded by a nucleic acid associated with PML, or a particular pattern of splicing variants.


Antibodies can be polyclonal or monoclonal and can be labeled or unlabeled. An intact antibody or a fragment thereof can be used. The term “labeled”, with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled as previously described herein. Other non-limiting examples of indirect labeling include detection of a primary antibody using a labeled secondary antibody, for example, a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled streptavidin.


Methods of Detecting Genetic Variations


In some embodiments, standard techniques for genotyping for the presence genetic variations, for example, amplification, can be used. Amplification of nucleic acids can be accomplished using methods known in the art. Generally, sequence information from the region of interest can be used to design oligonucleotide primers that can be identical or similar in sequence to opposite strands of a template to be amplified. In some embodiments, amplification methods can include but are not limited to, fluorescence-based techniques utilizing PCR, for example, ligase chain reaction (LCR), Nested PCR, transcription amplification, self-sustained sequence replication, nucleic acid based sequence amplification (NASBA), and multiplex ligation-dependent probe amplification (MLPA). Guidelines for selecting primers for PCR amplification are well known in the art. In some embodiments, a computer program can be used to design primers, for example, Oligo (National Biosciences, Inc, Plymouth Minn), MacVector (Kodak/IBI), and GCG suite of sequence analysis programs.


In some embodiments, commercial methodologies available for genotyping, for example, SNP genotyping, can be used, but are not limited to, TaqMan genotyping assays (Applied Biosystems), SNPlex platforms (Applied Biosystems), gel electrophoresis, capillary electrophoresis, size exclusion chromatography, mass spectrometry, for example, MassARRAY system (Sequenom), minisequencing methods, real-time Polymerase Chain Reaction (PCR), Bio-Plex system (BioRad), CEQ and SNPstream systems (Beckman), array hybridization technology, for example, Affymetrix GeneChip (Perlegen), BeadArray Technologies, for example, Illumina GoldenGate and Infinium assays, array tag technology, Multiplex Ligation-dependent Probe Amplification (MLPA), and endonuclease-based fluorescence hybridization technology (Invader assay, either using unamplified or amplified genomic DNA, or unamplified total RNA, or unamplified or amplified cDNA; Third Wave/Hologic). PCR can be a procedure in which target nucleic acid is amplified in a manner similar to that described in U.S. Pat. No. 4,683,195 and subsequent modifications of the procedure described therein. PCR can include a three phase temperature cycle of denaturation of DNA into single strands, annealing of primers to the denatured strands, and extension of the primers by a thermostable DNA polymerase enzyme. This cycle can be repeated so that there are enough copies to be detected and analyzed. In some embodiments, real-time quantitative PCR can be used to determine genetic variations, wherein quantitative PCR can permit both detection and quantification of a DNA sequence in a nucleic acid sample, for example, as an absolute number of copies or as a relative amount when normalized to DNA input or other normalizing genes. In some embodiments, methods of quantification can include the use of fluorescent dyes that can intercalate with double-stranded DNA, and modified DNA oligonucleotide probes that can fluoresce when hybridized with a complementary DNA.


In some embodiments of the disclosure, a nucleic acid sample obtained from the subject can be collected and PCR can used to amplify a fragment of nucleic acid that comprises one or more genetic variations that can be indicative of a susceptibility to PML. In some embodiments, detection of genetic variations can be accomplished by expression analysis, for example, by using quantitative PCR. In some embodiments, this technique can assess the presence or absence of a genetic alteration in the expression or composition of one or more polypeptides or splicing variants encoded by a nucleic acid associated with PML.


In some embodiments, the nucleic acid sample from a subject containing a SNP can be amplified by PCR prior to detection with a probe. In such an embodiment, the amplified DNA serves as the template for a detection probe and, in some embodiments, an enhancer probe. Certain embodiments of the detection probe, the enhancer probe, and/or the primers used for amplification of the template by PCR can comprise the use of modified bases, for example, modified A, T, C, G, and U, wherein the use of modified bases can be useful for adjusting the melting temperature of the nucleotide probe and/or primer to the template DNA, In some embodiments, modified bases are used in the design of the detection nucleotide probe. Any modified base known to the skilled person can be selected in these methods, and the selection of suitable bases is well within the scope of the skilled person based on the teachings herein and known bases available from commercial sources as known to the skilled person.


In some embodiments, identification of genetic variations can be accomplished using hybridization methods. The presence of a specific marker allele or a particular genomic segment comprising a genetic variation, or representative of a genetic variation, can be indicated by sequence-specific hybridization of a nucleic acid probe specific for the particular allele or the genetic variation in a nucleic acid sample that has or has not been amplified but methods described herein. The presence of more than one specific marker allele or several genetic variations can be indicated by using two or more sequence-specific nucleic acid probes, wherein each is specific for a particular allele and/or genetic variation.


Hybridization can be performed by methods well known to the person skilled in the art, for example, hybridization techniques such as fluorescent in situ hybridization (FISH), Southern analysis, Northern analysis, or in situ hybridization. In some embodiments, hybridization refers to specific hybridization, wherein hybridization can be performed with no mismatches. Specific hybridization, if present, can be using standard methods. In some embodiments, if specific hybridization occurs between a nucleic acid probe and the nucleic acid in the nucleic acid sample, the nucleic acid sample can contain a sequence that can be complementary to a nucleotide present in the nucleic acid probe. In some embodiments, if a nucleic acid probe can contain a particular allele of a polymorphic marker, or particular alleles for a plurality of markers, specific hybridization is indicative of the nucleic acid being completely complementary to the nucleic acid probe, including the particular alleles at polymorphic markers within the probe. In some embodiments a probe can contain more than one marker alleles of a particular haplotype, for example, a probe can contain alleles complementary to 2, 3, 4, 5 or all of the markers that make up a particular haplotype. In some embodiments detection of one or more particular markers of the haplotype in the nucleic acid sample is indicative that the source of the nucleic acid sample has the particular haplotype.


In some embodiments, PCR conditions and primers can be developed that amplify a product only when the variant allele is present or only when the wild type allele is present, for example, allele-specific PCR. In some embodiments of allele-specific PCR, a method utilizing a detection oligonucleotide probe comprising a fluorescent moiety or group at its 3′ terminus and a quencher at its 5′ terminus, and an enhancer oligonucleotide, can be employed (see e.g., Kutyavin et al., Nucleic Acid Res. 34:e128 (2006)).


An allele-specific primer/probe can be an oligonucleotide that is specific for particular a polymorphism can be prepared using standard methods. In some embodiments, allele-specific oligonucleotide probes can specifically hybridize to a nucleic acid region that contains a genetic variation. In some embodiments, hybridization conditions can be selected such that a nucleic acid probe can specifically bind to the sequence of interest, for example, the variant nucleic acid sequence.


In some embodiments, allele-specific restriction digest analysis can be used to detect the existence of a polymorphic variant of a polymorphism, if alternate polymorphic variants of the polymorphism can result in the creation or elimination of a restriction site. Allele-specific restriction digests can be performed, for example, with the particular restriction enzyme that can differentiate the alleles. In some embodiments, PCR can be used to amplify a region comprising the polymorphic site, and restriction fragment length polymorphism analysis can be conducted. In some embodiments, for sequence variants that do not alter a common restriction site, mutagenic primers can be designed that can introduce one or more restriction sites when the variant allele is present or when the wild type allele is present.


In some embodiments, fluorescence polarization template-directed dye-terminator incorporation (FP-TDI) can be used to determine which of multiple polymorphic variants of a polymorphism can be present in a subject. Unlike the use of allele-specific probes or primers, this method can employ primers that can terminate adjacent to a polymorphic site, so that extension of the primer by a single nucleotide can result in incorporation of a nucleotide complementary to the polymorphic variant at the polymorphic site.


In some embodiments, DNA containing an amplified portion can be dot-blotted, using standard methods and the blot contacted with the oligonucleotide probe. The presence of specific hybridization of the probe to the DNA can then be detected. The methods can include determining the genotype of a subject with respect to both copies of the polymorphic site present in the genome, wherein if multiple polymorphic variants exist at a site, this can be appropriately indicated by specifying which variants are present in a subject. Any of the detection means described herein can be used to determine the genotype of a subject with respect to one or both copies of the polymorphism present in the subject's genome.


In some embodiments, a peptide nucleic acid (PNA) probe can be used in addition to, or instead of, a nucleic acid probe in the methods described herein. A PNA can be a DNA mimic having a peptide-like, inorganic backbone, for example, N-(2-aminoethyl) glycine units with an organic base (A, G, C, T or U) attached to the glycine nitrogen via a methylene carbonyl linker.


Nucleic acid sequence analysis can also be used to detect genetic variations, for example, genetic variations can be detected by sequencing exons, introns, 5′ untranslated sequences, or 3′ untranslated sequences. One or more methods of nucleic acid analysis that are available to those skilled in the art can be used to detect genetic variations, including but not limited to, direct manual sequencing, automated fluorescent sequencing, single-stranded conformation polymorphism assays (SSCP); clamped denaturing gel electrophoresis (CDGE); denaturing gradient gel electrophoresis (DGGE), two-dimensional gel electrophoresis (2DGE or TDGE); conformational sensitive gel electrophoresis (CSGE); denaturing high performance liquid chromatography (DHPLC), infrared matrix-assisted laser desorption/ionization (IR-MALDI) mass spectrometry, mobility shift analysis, quantitative real-time PCR, restriction enzyme analysis, heteroduplex analysis; chemical mismatch cleavage (CMC), RNase protection assays, use of polypeptides that recognize nucleotide mismatches, allele-specific PCR, real-time pyrophosphate DNA sequencing, PCR amplification in combination with denaturing high performance liquid chromatography (dHPLC), and combinations of such methods.


Sequencing can be accomplished through classic Sanger sequencing methods, which are known in the art. In some embodiments sequencing can be performed using high-throughput sequencing methods some of which allow detection of a sequenced nucleotide immediately after or upon its incorporation into a growing strand, for example, detection of sequence in substantially real time or real time. In some cases, high throughput sequencing generates at least 1,000, at least 5,000, at least 10,000, at least 20,000, at least 30,000, at least 40,000, at least 50,000, at least 100,000 or at least 500,000 sequence reads per hour; with each read being at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 120 or at least 150 bases per read (or 500-1,000 bases per read for 454).


High-throughput sequencing methods can include but are not limited to, Massively Parallel Signature Sequencing (MPSS, Lynx Therapeutics), Polony sequencing, 454 pyrosequencing, Illumina (Solexa) sequencing, Illumina (Solexa) sequencing using 10× Genomics library preparation, SOLiD sequencing, on semiconductor sequencing, DNA nanoball sequencing, Helioscope™ single molecule sequencing, Single Molecule SMRT™ sequencing, Single Molecule real time (RNAP) sequencing, Nanopore DNA sequencing, and/or sequencing by hybridization, for example, a non-enzymatic method that uses a DNA microarray, or microfluidic Sanger sequencing.


In some embodiments, high-throughput sequencing can involve the use of technology available by Helicos BioSciences Corporation (Cambridge, Mass.) such as the Single Molecule Sequencing by Synthesis (SMSS) method. SMSS is unique because it allows for sequencing the entire human genome in up to 24 hours. This fast sequencing method also allows for detection of a SNP/nucleotide in a sequence in substantially real time or real time. Finally, SMSS is powerful because, like the MIP technology, it does not use a pre-amplification step prior to hybridization. SMSS does not use any amplification. SMSS is described in US Publication Application Nos. 20060024711; 20060024678; 20060012793; 20060012784; and 20050100932. In some embodiments, high-throughput sequencing involves the use of technology available by 454 Life Sciences, Inc. (a Roche company, Branford, Conn.) such as the PicoTiterPlate device which includes a fiber optic plate that transmits chemiluminescent signal generated by the sequencing reaction to be recorded by a CCD camera in the instrument. This use of fiber optics allows for the detection of a minimum of 20 million base pairs in 4.5 hours.


In some embodiments, PCR-amplified single-strand nucleic acid can be hybridized to a primer and incubated with a polymerase, ATP sulfurylase, luciferase, apyrase, and the substrates luciferin and adenosine 5′ phosphosulfate. Next, deoxynucleotide triphosphates corresponding to the bases A, C, G, and T (U) can be added sequentially. A base incorporation can be accompanied by release of pyrophosphate, which can be converted to ATP by sulfurylase, which can drive synthesis of oxyluciferin and the release of visible light. Since pyrophosphate release can be equimolar with the number of incorporated bases, the light given off can be proportional to the number of nucleotides adding in any one step. The process can repeat until the entire sequence can be determined. In some embodiments, pyrosequencing can be utilized to analyze amplicons to determine whether breakpoints are present. In some embodiments, pyrosequencing can map surrounding sequences as an internal quality control.


Pyrosequencing analysis methods are known in the art. Sequence analysis can include a four-color sequencing by ligation scheme (degenerate ligation), which involves hybridizing an anchor primer to one of four positions. Then an enzymatic ligation reaction of the anchor primer to a population of degenerate nonamers that are labeled with fluorescent dyes can be performed. At any given cycle, the population of nonamers that is used can be structured such that the identity of one of its positions can be correlated with the identity of the fluorophore attached to that nonamer. To the extent that the ligase discriminates for complementarily at that queried position, the fluorescent signal can allow the inference of the identity of the base. After performing the ligation and four-color imaging, the anchor primer: nonamer complexes can be stripped and a new cycle begins. Methods to image sequence information after performing ligation are known in the art.


In some embodiments, analysis by restriction enzyme digestion can be used to detect a particular genetic variation if the genetic variation results in creation or elimination of one or more restriction sites relative to a reference sequence. In some embodiments, restriction fragment length polymorphism (RFLP) analysis can be conducted, wherein the digestion pattern of the relevant DNA fragment indicates the presence or absence of the particular genetic variation in the nucleic acid sample.


In some embodiments, arrays of oligonucleotide probes that can be complementary to target nucleic acid sequence segments from a subject can be used to identify genetic variations. In some embodiments, an array of oligonucleotide probes comprises an oligonucleotide array, for example, a microarray. In some embodiments, the present disclosure features arrays that include a substrate having a plurality of addressable areas, and methods of using them. At least one area of the plurality includes a nucleic acid probe that binds specifically to a sequence comprising a genetic variation, and can be used to detect the absence or presence of the genetic variation, for example, one or more SNPs, microsatellites, or CNVs, as described herein, to determine or identify an allele or genotype. For example, the array can include one or more nucleic acid probes that can be used to detect a genetic variation associated with a gene and/or gene product. In some embodiments, the array can further comprise at least one area that includes a nucleic acid probe that can be used to specifically detect another marker associated with PML as described herein.


Microarray hybridization can be performed by hybridizing a nucleic acid of interest, for example, a nucleic acid encompassing a genetic variation, with the array and detecting hybridization using nucleic acid probes. In some embodiments, the nucleic acid of interest is amplified prior to hybridization. Hybridization and detecting can be carried out according to standard methods described in Published PCT Applications: WO 92/10092 and WO 95/11995, and U.S. Pat. No. 5,424,186. For example, an array can be scanned to determine the position on the array to which the nucleic acid hybridizes. The hybridization data obtained from the scan can be, for example, in the form of fluorescence intensities as a function of location on the array.


Arrays can be formed on substrates fabricated with materials such as paper; glass; plastic, for example, polypropylene, nylon, or polystyrene; polyacrylamide; nitrocellulose; silicon; optical fiber; or any other suitable solid or semisolid support; and can be configured in a planar, for example, glass plates or silicon chips); or three dimensional, for example, pins, fibers, beads, particles, microtiter wells, and capillaries, configuration.


Methods for generating arrays are known in the art and can include for example; photolithographic methods (U.S. Pat. Nos. 5,143,854, 5,510,270 and 5,527,681); mechanical methods, for example, directed-flow methods (U.S. Pat. No. 5,384,261); pin-based methods (U.S. Pat. No. 5,288,514); bead-based techniques (PCT US/93/04145); solid phase oligonucleotide synthesis methods; or by other methods known to a person skilled in the art (see, e.g., Bier, F. F., et al., Adv Biochem Eng Biotechnol 109:433-53 (2008); Hoheisel, J. D., Nat Rev Genet 7: 200-10 (2006); Fan, J. B., et al., Methods Enzymol 410:57-73 (2006); Raqoussis, J. & Elvidge, G., Expert Rev Mol Design 6: 145-52 (2006); Mockler, T. C., et al., Genomics 85: 1-15 (2005), and references cited therein, the entire teachings of each of which are incorporated by reference herein). Many additional descriptions of the preparation and use of oligonucleotide arrays for detection of polymorphisms can be found, for example, in U.S. Pat. Nos. 6,858,394, 6,429,027, 5,445,934, 5,700,637, 5,744,305, 5,945,334, 6,054,270, 6,300,063, 6,733,977, 7,364,858, EP 619 321, and EP 373 203, the entire teachings of which are incorporated by reference herein. Methods for array production, hybridization, and analysis are also described in Snijders et al., Nat. Genetics 29:263-264 (2001); Klein et al., Proc. Natl. Acad. Sci. USA 96:4494-4499 (1999); Albertson et al., Breast Cancer Research and Treatment 78:289-298 (2003); and Snijders et al., “BAC microarray based comparative genomic hybridization,” in: Zhao et al., (eds), Bacterial Artificial Chromosomes: Methods and Protocols, Methods in Molecular Biology, Humana Press (2002).


In some embodiments, oligonucleotide probes forming an array can be attached to a substrate by any number of techniques, including, but not limited to, in situ synthesis, for example, high-density oligonucleotide arrays, using photolithographic techniques; spotting/printing a medium to low density on glass, nylon, or nitrocellulose; by masking; and by dot-blotting on a nylon or nitrocellulose hybridization membrane. In some embodiments, oligonucleotides can be immobilized via a linker, including but not limited to, by covalent, ionic, or physical linkage. Linkers for immobilizing nucleic acids and polypeptides, including reversible or cleavable linkers, are known in the art (U.S. Pat. No. 5,451,683 and WO98/20019). In some embodiments, oligonucleotides can be non-covalently immobilized on a substrate by hybridization to anchors, by means of magnetic beads, or in a fluid phase, for example, in wells or capillaries.


An array can comprise oligonucleotide hybridization probes capable of specifically hybridizing to different genetic variations. In some embodiments, oligonucleotide arrays can comprise a plurality of different oligonucleotide probes coupled to a surface of a substrate in different known locations. In some embodiments, oligonucleotide probes can exhibit differential or selective binding to polymorphic sites, and can be readily designed by one of ordinary skill in the art, for example, an oligonucleotide that is perfectly complementary to a sequence that encompasses a polymorphic site, for example, a sequence that includes the polymorphic site, within it, or at one end, can hybridize preferentially to a nucleic acid comprising that sequence, as opposed to a nucleic acid comprising an alternate polymorphic variant.


In some embodiments, arrays can include multiple detection blocks, for example, multiple groups of probes designed for detection of particular polymorphisms. In some embodiments, these arrays can be used to analyze multiple different polymorphisms. In some embodiments, detection blocks can be grouped within a single array or in multiple, separate arrays, wherein varying conditions, for example, conditions optimized for particular polymorphisms, can be used during hybridization. General descriptions of using oligonucleotide arrays for detection of polymorphisms can be found, for example, in U.S. Pat. Nos. 5,858,659 and 5,837,832. In addition to oligonucleotide arrays, cDNA arrays can be used similarly in certain embodiments.


The methods described herein can include but are not limited to providing an array as described herein; contacting the array with a nucleic acid sample, and detecting binding of a nucleic acid from the nucleic acid sample to the array. In some embodiments, the method can comprise amplifying nucleic acid from the nucleic acid sample, for example, a region associated with PML or a region that includes another region associated with PML. In some embodiments, the methods described herein can include using an array that can identify differential expression patterns or copy numbers of one or more genes in nucleic acid samples from control and affected individuals. For example, arrays of probes to a marker described herein can be used to identify genetic variations between DNA from an affected subject, and control DNA obtained from an individual that does not have PML. Since the nucleotides on the array can contain sequence tags, their positions on the array can be accurately known relative to the genomic sequence.


In some embodiments, it can be desirable to employ methods that can detect the presence of multiple genetic variations, for example, polymorphic variants at a plurality of polymorphic sites, in parallel or substantially simultaneously. In some embodiments, these methods can comprise oligonucleotide arrays and other methods, including methods in which reactions, for example, amplification and hybridization, can be performed in individual vessels, for example, within individual wells of a multi-well plate or other vessel.


Determining the identity of a genetic variation can also include or consist of reviewing a subject's medical history, where the medical history includes information regarding the identity, copy number, presence or absence of one or more alleles or SNPs in the subject, e.g., results of a genetic test.


In some embodiments extended runs of homozygosity (ROH) may be useful to map recessive disease genes in outbred populations. Furthermore, even in complex disorders, a high number of affected individuals may have the same haplotype in the region surrounding a disease mutation. Therefore, a rare pathogenic variant and surrounding haplotype can be enriched in frequency in a group of affected individuals compared with the haplotype frequency in a cohort of unaffected controls. Homozygous haplotypes (HH) that are shared by multiple affected individuals can be important for the discovery of recessive disease genes in a condition such as PML. In some embodiments, the traditional homozygosity mapping method can be extended by analyzing the haplotype within shared ROH regions to identify homozygous segments of identical haplotype that are present uniquely or at a higher frequency in PML probands compared to parental controls. Such regions are termed risk homozygous haplotypes (rHH), which may contain low-frequency recessive variants that contribute to PML risk in a subset of PML patients.


Genetic variations can also be identified using any of a number of methods well known in the art. For example, genetic variations available in public databases, which can be searched using methods and custom algorithms or algorithms known in the art, can be used. In some embodiments, a reference sequence can be from, for example, the human draft genome sequence, publicly available in various databases, or a sequence deposited in a database such as GenBank.


A comparison of one or more genomes relative to one or more other genomes with array CGH, or a variety of other genetic variation detection methods, can reveal the set of genetic variations between two genomes, between one genome in comparison to multiple genomes, or between one set of genomes in comparison to another set of genomes. In some embodiments, an array CGH experiment can be performed by hybridizing a single test genome against a pooled nucleic acid sample of two or more genomes, which can result in minimizing the detection of higher frequency variants in the experiment. In some embodiments, a test genome can be hybridized alone (i.e., one-color detection) to a microarray, for example, using array CGH or SNP genotyping methods, and the comparison step to one or more reference genomes can be performed in silico to reveal the set of genetic variations in the test genome relative to the one or more reference genomes. In one embodiment, a single test genome is compared to a single reference genome in a 2-color experiment wherein both genomes are cohybridized to the microarray. In some embodiments, the whole genome or whole exome from one or more subjects is analyzed. In some embodiments, nucleic acid information has already been obtained for the whole genome or whole exome from one or more individuals and the nucleic acid information is obtained from in silico analysis.


Any of the polynucleotides described, including polynucleotides comprising a genetic variation, can be made synthetically using methods known in the art.


Methods of Detecting CNVs


Detection of genetic variations, specifically CNVs, can be accomplished by one or more suitable techniques described herein. Generally, techniques that can selectively determine whether a particular chromosomal segment is present or absent in an individual can be used for genotyping CNVs. Identification of novel copy number variations can be done by methods for assessing genomic copy number changes.


In some embodiments, methods include but are not limited to, methods that can quantitatively estimate the number of copies of a particular genomic segment, but can also include methods that indicate whether a particular segment is present in a nucleic acid sample or not. In some embodiments, the technique to be used can quantify the amount of segment present, for example, determining whether a DNA segment is deleted, duplicated, or triplicated in subject, for example, Fluorescent In Situ Hybridization (FISH) techniques, and other methods described herein. In some embodiments, methods include detection of copy number variation from array intensity and sequencing read depth using a stepwise Bayesian model (Zhang, et al., BMC Bioinformatics, 11:539 (2010)). In some embodiments, methods include detecting copy number variations using shotgun sequencing, CNV-seq (Xie C., et al., BMC Bioinformatics, 10:80 (2009)). In some embodiments, methods include analyzing next-generation sequencing (NGS) data for CNV detection using any one of several algorithms developed for each of the four broad methods for CNV detection using NGS, namely the depth of coverage (DOC), read-pair (RP), split-read (SR) and assembly-based (AS) methods. (Teo et al., Bioinformatics (2012)). In some embodiments, methods include combining coverage with map information for the identification of deletions and duplications in targeted sequence data (Nord et al., BMC Genomics, 12:184 (2011)).


In some embodiments, other genotyping technologies can be used for detection of CNVs, including but not limited to, karyotype analysis, Molecular Inversion Probe array technology, for example, Affymetrix SNP Array 6.0, and BeadArray Technologies, for example, Illumina GoldenGate and Infinium assays, as can other platforms such as NimbleGen HD2.1 or HD4.2, High-Definition Comparative Genomic Hybridization (CGH) arrays (Agilent Technologies), tiling array technology (Affymetrix), multiplex ligation-dependent probe amplification (MLPA), Invader assay, fluorescence in situ hybridization, and, in one embodiment, Array Comparative Genomic Hybridization (aCGH) methods. As described herein, karyotype analysis can be a method to determine the content and structure of chromosomes in a nucleic acid sample. In some embodiments, karyotyping can be used, in lieu of aCGH, to detect translocations or inversions, which can be copy number neutral, and, therefore, not detectable by aCGH. Information about amplitude of particular probes, which can be representative of particular alleles, can provide quantitative dosage information for the particular allele, and by consequence, dosage information about the CNV in question, since the marker can be selected as a marker representative of the CNV and can be located within the CNV. In some embodiments, if the CNV is a deletion, the absence of particular marker allele is representative of the deletion. In some embodiments, if the CNV is a duplication or a higher order copy number variation, the signal intensity representative of the allele correlating with the CNV can represent the copy number. A summary of methodologies commonly used is provided in Perkel (Perkel J. Nature Methods 5:447-453 (2008)).


PCR assays can be utilized to detect CNVs and can provide an alternative to array analysis. In particular, PCR assays can enable detection of precise boundaries of gene/chromosome variants, at the molecular level, and which boundaries are identical in different individuals. PCR assays can be based on the amplification of a junction fragment present only in individuals that carry a deletion. This assay can convert the detection of a loss by array CGH to one of a gain by PCR.


Examples of PCR techniques that can be used in the present disclosure include, but are not limited to quantitative PCR, real-time quantitative PCR (qPCR), quantitative fluorescent PCR (QF-PCR), multiplex fluorescent PCR (MF-PCR), real time PCR (RT-PCR), single cell PCR, PCR-RFLP/RT-PCR-RFLP, hot start PCR and Nested PCR. Other suitable amplification methods include the ligase chain reaction (LCR), ligation mediated PCR (LM-PCR), degenerate oligonucleotide probe PCR (DOP-PCR), transcription amplification, self-sustained sequence replication, selective amplification of target polynucleotide sequences, consensus sequence primed polymerase chain reaction (CP-PCR), arbitrarily primed polymerase chain reaction (AP-PCR) and nucleic acid sequence based amplification (NASBA).


Alternative methods for the simultaneous interrogation of multiple regions include quantitative multiplex PCR of short fluorescent fragments (QMPSF), multiplex amplifiable probe hybridization (MAPH) and multiplex ligation-dependent probe amplification (MLPA), in which copy-number differences for up to 40 regions can be scored in one experiment. Another approach can be to specifically target regions that harbor known segmental duplications, which are often sites of copy-number variation. By targeting the variable nucleotides between two copies of a segmental duplication (called paralogous sequence variants) using a SNP-genotyping method that provides independent fluorescence intensities for the two alleles, it is possible to detect an increase in intensity of one allele compared with the other.


In some embodiments, the amplified piece of DNA can be bound to beads using the sequencing element of the nucleic acid tag under conditions that favor a single amplified piece of DNA molecule to bind a different bead and amplification occurs on each bead. In some embodiments, such amplification can occur by PCR. Each bead can be placed in a separate well, which can be a picoliter-sized well. In some embodiments, each bead is captured within a droplet of a PCR-reaction-mixture-in-oil-emulsion and PCR amplification occurs within each droplet. The amplification on the bead results in each bead carrying at least one million, at least 5 million, or at least 10 million copies of the single amplified piece of DNA molecule.


In embodiments where PCR occurs in oil-emulsion mixtures, the emulsion droplets are broken, the DNA is denatured and the beads carrying single-stranded nucleic acids clones are deposited into a well, such as a picoliter-sized well, for further analysis according to the methods described herein. These amplification methods allow for the analysis of genomic DNA regions. Methods for using bead amplification followed by fiber optics detection are described in Margulies et al., Nature, 15; 437(7057):376-80 (2005), and as well as in US Publication Application Nos. 20020012930; 20030068629; 20030100102; 20030148344; 20040248161; 20050079510, 20050124022; and 20060078909.


Another variation on the array-based approach can be to use the hybridization signal intensities that are obtained from the oligonucleotides employed on Affymetrix SNP arrays or in Illumina Bead Arrays. Here hybridization intensities are compared with average values that are derived from controls, such that deviations from these averages indicate a change in copy number. As well as providing information about copy number, SNP arrays have the added advantage of providing genotype information. For example, they can reveal loss of heterozygosity, which could provide supporting evidence for the presence of a deletion, or might indicate segmental uniparental disomy (which can recapitulate the effects of structural variation in some genomic regions—Prader-Willi and Angelman syndromes, for example).


Many of the basic procedures followed in microarray-based genome profiling are similar, if not identical, to those followed in expression profiling and SNP analysis, including the use of specialized microarray equipment and data-analysis tools. Since microarray-based expression profiling has been well established in the last decade, much can be learned from the technical advances made in this area. Examples of the use of microarrays in nucleic acid analysis that can be used are described in U.S. Pat. Nos. 6,300,063, 5,837,832, 6,969,589, 6,040,138, 6,858,412, U.S. application Ser. No. 08/529,115, U.S. application Ser. No. 10/272,384, U.S. application Ser. No. 10/045,575, U.S. application Ser. No. 10/264,571 and U.S. application Ser. No. 10/264,574. It should be noted that there are also distinct differences such as target and probe complexity, stability of DNA over RNA, the presence of repetitive DNA and the need to identify single copy number alterations in genome profiling.


In some embodiments, the genetic variations detected comprise CNVs and can be detected using array CGH. In some embodiments, array CGH can be been implemented using a wide variety of techniques. The initial approaches used arrays produced from large-insert genomic clones such as bacterial artificial chromosomes (BACs). Producing sufficient BAC DNA of adequate purity to make arrays is arduous, so several techniques to amplify small amounts of starting material have been employed. These techniques include ligation-mediated PCR (Snijders et al., Nat. Genet. 29:263-64), degenerate primer PCR using one or several sets of primers, and rolling circle amplification. BAC arrays that provide complete genome tiling paths are also available. Arrays made from less complex nucleic acids such as cDNAs, selected PCR products, and oligonucleotides can also be used. Although most CGH procedures employ hybridization with total genomic DNA, it is possible to use reduced complexity representations of the genome produced by PCR techniques. Computational analysis of the genome sequence can be used to design array elements complementary to the sequences contained in the representation. Various SNP genotyping platforms, some of which use reduced complexity genomic representations, can be useful for their ability to determine both DNA copy number and allelic content across the genome. In some embodiments, small amounts of genomic DNA can be amplified with a variety of whole genome or whole exome amplification methods prior to CGH analysis of the nucleic acid sample. A “whole exome,” as used herein, includes exons throughout the whole genome that are expressed in genes. Since exon selection has tissue and cell type specificity, these positions may be different in the various cell types resulting from a splice variant or alternative splicing. A “whole genome,” as used herein, includes the entire genetic code of a genome.


The different basic approaches to array CGH provide different levels of performance, so some are more suitable for particular applications than others. The factors that determine performance include the magnitudes of the copy number changes, their genomic extents, the state and composition of the specimen, how much material is available for analysis, and how the results of the analysis can be used. Many applications use reliable detection of copy number changes of much less than 50%, a more stringent requirement than for other microarray technologies. Note that technical details are extremely important and different implementations of methods using the same array CGH approach can yield different levels of performance. Various CGH methods are known in the art and are equally applicable to one or more methods of the present disclosure. For example, CGH methods are disclosed in U.S. Pat. Nos. 7,030,231; 7,011,949; 7,014,997; 6,977,148; 6,951,761; and 6,916,621, the disclosure from each of which is incorporated by reference herein in its entirety.


The data provided by array CGH are quantitative measures of DNA sequence dosage. Array CGH provides high-resolution estimates of copy number aberrations, and can be performed efficiently on many nucleic acid samples. The advent of array CGH technology makes it possible to monitor DNA copy number changes on a genomic scale and many projects have been launched for studying the genome in specific diseases.


In some embodiments, whole genome array-based comparative genome hybridization (array CGH) analysis, or array CGH on a subset of genomic regions, can be used to efficiently interrogate human genomes for genomic imbalances at multiple loci within a single assay. The development of comparative genomic hybridization (CGH) (Kallioniemi et al., Science 258: 818-21 (1992)) provided the first efficient approach to scanning entire genomes for variations in DNA copy number. The importance of normal copy number variation involving large segments of DNA has been unappreciated. Array CGH is a breakthrough technique in human genetics, which is attracting interest from clinicians working in fields as diverse as cancer and IVF (In Vitro Fertilization). The use of CGH microarrays in the clinic holds great promise for identifying regions of genomic imbalance associated with disease. Advances from identifying chromosomal critical regions associated with specific phenotypes to identifying the specific dosage sensitive genes can lead to therapeutic opportunities of benefit to patients. Array CGH is a specific, sensitive and rapid technique that can enable the screening of the whole genome in a single test. It can facilitate and accelerate the screening process in human genetics and is expected to have a profound impact on the screening and counseling of patients with genetic disorders. It is now possible to identify the exact location on the chromosome where an aberration has occurred and it is possible to map these changes directly onto the genomic sequence.


An array CGH approach provides a robust method for carrying out a genome-wide scan to find novel copy number variants (CNVs). The array CGH methods can use labeled fragments from a genome of interest, which can be competitively hybridized with a second differentially labeled genome to arrays that are spotted with cloned DNA fragments, revealing copy-number differences between the two genomes. Genomic clones (for example, BACs), cDNAs, PCR products and oligonucleotides, can all be used as array targets. The use of array CGH with BACs was one of the earliest employed methods and is popular, owing to the extensive coverage of the genome it provides, the availability of reliable mapping data and ready access to clones. The last of these factors is important both for the array experiments themselves, and for confirmatory FISH experiments.


In a typical CGH measurement, total genomic DNA is isolated from control and reference subjects, differentially labeled, and hybridized to a representation of the genome that allows the binding of sequences at different genomic locations to be distinguished. More than two genomes can be compared simultaneously with suitable labels. Hybridization of highly repetitive sequences is typically suppressed by the inclusion of unlabeled Cot-1 DNA in the reaction. In some embodiments of array CGH, it is beneficial to mechanically shear the genomic DNA in a nucleic acid sample, for example, with sonication, prior to its labeling and hybridization step. In another embodiment, array CGH may be performed without use of Cot-1 DNA or a sonication step in the preparation of the genomic DNA in a nucleic acid sample. The relative hybridization intensity of the test and reference signals at a given location can be proportional to the relative copy number of those sequences in the test and reference genomes. If the reference genome is normal then increases and decreases in signal intensity ratios directly indicate DNA copy number variation within the genome of the test cells. Data are typically normalized so that the modal ratio for the genome is set to some standard value, typically 1.0 on a linear scale or 0.0 on a logarithmic scale. Additional measurements such as FISH or flow cytometry can be used to determine the actual copy number associated with a ratio level.


In some embodiments, an array CGH procedure can include the following steps. First, large-insert clones, for example, BACs can be obtained from a supplier of clone libraries. Then, small amounts of clone DNA can be amplified, for example, by degenerate oligonucleotide-primed (DOP) PCR or ligation-mediated PCR in order to obtain sufficient quantities needed for spotting. Next, PCR products can be spotted onto glass slides using, for example, microarray robots equipped with high-precision printing pins. Depending on the number of clones to be spotted and the space available on the microarray slide, clones can either be spotted once per array or in replicate. Repeated spotting of the same clone on an array can increase precision of the measurements if the spot intensities are averaged, and allows for a detailed statistical analysis of the quality of the experiments. Subject and control DNAs can be labeled, for example, with either Cy3 or Cy5-dUTP using random priming and can be subsequently hybridized onto the microarray in a solution containing an excess of Cot1-DNA to block repetitive sequences. Hybridizations can either be performed manually under a coverslip, in a gasket with gentle rocking or, automatically using commercially available hybridization stations. These automated hybridization stations can allow for an active hybridization process, thereby improving the reproducibility as well as reducing the actual hybridization time, which increases throughput. The hybridized DNAs can detected through the two different fluorochromes using standard microarray scanning equipment with either a scanning confocal laser or a charge coupled device (CCD) camera-based reader, followed by spot identification using commercially or freely available software packages.


The use of CGH with arrays that comprise long oligonucleotides (60-100 bp) can improve the detection resolution (in some embodiments, as small as ˜3-5 kb sized CNVs on arrays designed for interrogation of human whole genomes) over that achieved using BACs (limited to 50-100 kb or larger sized CNVs due to the large size of BAC clones). In some embodiments, the resolution of oligonucleotide CGH arrays is achieved via in situ synthesis of 1-2 million unique features/probes per microarray, which can include microarrays available from Roche NimbleGen and Agilent Technologies. In addition to array CGH methods for copy number detection, other embodiments for partial or whole genome analysis of CNVs within a genome include, but are not limited to, use of SNP genotyping microarrays and sequencing methods.


Another method for copy number detection that uses oligonucleotides can be representational oligonucleotide microarray analysis (ROMA). It is similar to that applied in the use of BAC and CGH arrays, but to increase the signal-to-noise ratio, the ‘complexity’ of the input DNA is reduced by a method called representation or whole-genome sampling. Here the DNA that is to be hybridized to the array can be treated by restriction digestion and then ligated to adapters, which results in the PCR-based amplification of fragments in a specific size-range. As a result, the amplified DNA can make up a fraction of the entire genomic sequence—that is, it is a representation of the input DNA that has significantly reduced complexity, which can lead to a reduction in background noise. Other suitable methods available to the skilled person can also be used, and are within scope of the present disclosure.


A comparison of one or more genomes relative to one or more other genomes with array CGH, or a variety of other CNV detection methods, can reveal the set of CNVs between two genomes, between one genome in comparison to multiple genomes, or between one set of genomes in comparison to another set of genomes. In some embodiments, an array CGH experiment can be performed by hybridizing a single test genome against a pooled nucleic acid sample of two or more genomes, which can result in minimizing the detection of higher frequency variants in the experiment. In some embodiments, a test genome can be hybridized alone (i.e. one-color detection) to a microarray, for example, using array CGH or SNP genotyping methods, and the comparison step to one or more reference genomes can be performed in silico to reveal the set of CNVs in the test genome relative to the one or more reference genomes. In one preferred embodiment, a single test genome is compared to a single reference genome in a 2-color experiment wherein both genomes are cohybridized to the microarray.


Array CGH can be used to identify genes that are causative or associated with a particular phenotype, condition, or disease by comparing the set of CNVs found in the affected cohort to the set of CNVs found in an unaffected cohort. An unaffected cohort may consist of any individual unaffected by the phenotype, condition, or disease of interest, but in one preferred embodiment is comprised of individuals or subjects that are apparently healthy (normal). Methods employed for such analyses are described in U.S. Pat. Nos. 7,702,468 and 7,957,913. In some embodiments of CNV comparison methods, candidate genes that are causative or associated (i.e. potentially serving as a biomarker) with a phenotype, condition, or disease will be identified by CNVs that occur in the affected cohort but not in the unaffected cohort. In some embodiments of CNV comparison methods, candidate genes that are causative or associated (i.e. potentially serving as a biomarker) with a phenotype, condition, or disease will be identified by CNVs that occur at a statistically significant higher frequency in the affected cohort as compared their frequency in the unaffected cohort. Thus, CNVs preferentially detected in the affected cohort as compared to the unaffected cohort can serve as beacons of genes that are causative or associated with a particular phenotype, condition, or disease. Methods employed for such analyses are described in U.S. Pat. No. 8,862,410. In some embodiments, CNV detection and comparison methods can result in direct identification of the gene that is causative or associated with phenotype, condition, or disease if the CNVs are found to overlap with or encompass the gene(s). In some embodiments, CNV detection and comparison methods can result in identification of regulatory regions of the genome (e.g., promoters, enhancers, transcription factor binding sites) that regulate the expression of one or more genes that are causative or associated with the phenotype, condition, or disease of interest. In some embodiments, CNV detection and comparison methods can result in identification of a region in the genome in linkage disequilibrium with a genetic variant that is causative or associated with the phenotype, condition, or disease of interest. In another embodiment, CNV detection and comparison methods can result in identification of a region in the genome in linkage disequilibrium with a genetic variant that is protective against the condition or disease of interest.


Due to the large amount of genetic variation between any two genomes, or two sets (cohorts) of genomes, being compared, one preferred embodiment is to reduce the genetic variation search space by interrogating only CNVs, as opposed to the full set of genetic variants that can be identified in an individual's genome or exome. The set of CNVs that occur only, or at a statistically higher frequency, in the affected cohort as compared to the unaffected cohort can then be further investigated in targeted sequencing experiments to reveal the full set of genetic variants (of any size or type) that are causative or associated (i.e. potentially serving as a biomarker) with a phenotype, condition, or disease. It can be appreciated to those skilled in the art that the targeted sequencing experiments are performed in both the affected and unaffected cohorts in order to identify the genetic variants (e.g., SNVs and indels) that occur only, or at a statistically significant higher frequency, in the affected individual or cohort as compared to the unaffected cohort. Methods employed for such analyses are described in U.S. Pat. No. 8,862,410.


A method of screening a subject for a disease or disorder can comprise assaying a nucleic acid sample from the subject to detect sequence information for more than one genetic locus and comparing the sequence information to a panel of nucleic acid biomarkers and screening the subject for the presence or absence of the disease or disorder if one or more of low frequency biomarkers in the panel are present in the sequence information.


The panel can comprise at least one nucleic acid biomarker for each of the more than one genetic loci. For example, the panel can comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 150, 200 or more nucleic acid biomarkers for each of the more than one genetic locus. In some embodiments, the panel can comprise from about 2-1000 nucleic acid biomarkers. For example, the panel can comprise from about 2-900, 2-800, 2-700, 2-600, 2-500, 2-400, 2-300, 2-200, 2-100, 25-900, 25-800, 25-700, 25-600, 25-500, 25-400, 25-300, 25-200, 25-100, 100-1000, 100-900, 100-800, 100-700, 100-600, 100-500, 100-400, 100-300, 100-200, 200-1000, 200-900, 200-800, 200-700, 200-600, 200-500, 200-400, 200-300, 300-1000, 300-900, 300-800, 300-700, 300-600, 300-500, 300-400, 400-1000, 400-900, 400-800, 400-700, 400-600, 400-500, 500-1000, 500-900, 500-800, 500-700, 500-600, 600-1000, 600-900, 600-800, 600-700, 700-1000, 700-900, 700-800, 800-1000, 800-900, or 900-1000 nucleic acid biomarkers.


In some embodiments, a biomarker can occur at a frequency of 1% or more in a population of subjects without a diagnosis of the disease or disorder. For example, a biomarker can occur at a frequency of 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or more in a population of subjects without a diagnosis of the disease or disorder. In some embodiments, a biomarker can occur at a frequency from about 1%-20% in a population of subjects without a diagnosis of the disease or disorder. For example, a biomarker can occur at a frequency of from about 1%-5% or 1%-10%, in a population of subjects without a diagnosis of the disease or disorder.


The panel can comprise at least 2 low frequency biomarkers. For example, the panel can comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 3, 14, 15, 15, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 250, 500, or 1000 or more low frequency biomarkers. In some embodiments, the panel can comprise from about 2-1000 low frequency biomarkers. For example, the panel can comprise from about 2-900, 2-800, 2-700, 2-600, 2-500, 2-400, 2-300, 2-200, 2-100, 25-900, 25-800, 25-700, 25-600, 25-500, 25-400, 25-300, 25-200, 25-100, 100-1000, 100-900, 100-800, 100-700, 100-600, 100-500, 100-400, 100-300, 100-200, 200-1000, 200-900, 200-800, 200-700, 200-600, 200-500, 200-400, 200-300, 300-1000, 300-900, 300-800, 300-700, 300-600, 300-500, 300-400, 400-1000, 400-900, 400-800, 400-700, 400-600, 400-500, 500-1000, 500-900, 500-800, 500-700, 500-600, 600-1000, 600-900, 600-800, 600-700, 700-1000, 700-900, 700-800, 800-1000, 800-900, or 900-1000 low frequency biomarkers.


In some embodiments, a low frequency biomarker can occur at a frequency of 1% or less in a population of subjects without a diagnosis of the disease or disorder. For example, a low frequency biomarker can occur at a frequency of 0.5%, 0.1%, 0.05%, 0.01%, 0.005%, 0.001%, 0.0005%, or 0.0001% or less in a population of subjects without a diagnosis of the disease or disorder. In some embodiments, a low frequency biomarker can occur at a frequency from about 0.0001%-0.1% in a population of subjects without a diagnosis of the disease or disorder. For example, a low frequency biomarker can occur at a frequency of from about 0.0001%-0.0005%, 0.0001%-0.001%, 0.0001%-0.005%, 0.0001%-0.01%, 0.0001%-0.05%, 0.0001%-0.1%, 0.0001%-0.5%, 0.0005%-0.001%, 0.0005%-0.005%, 0.0005%-0.01%, 0.0005%-0.05%, 0.0005%-0.1%, 0.0005%-0.5%, 0.0005%-1%, 0.001%-0.005%, 0.001%-0.01%, 0.001%-0.05%, 0.001%-0.1%, 0.001%-0.5%, 0.001%-1%, 0.005%-0.01%, 0.005%-0.05%, 0.005%-0.1%, 0.005%-0.5%, 0.005%-1%, 0.01%-0.05%, 0.01%-0.1%, 0.01%-0.5%, 0.01%-1%, 0.05%-0.1%, 0.05%-0.5%, 0.05%-1%, 0.1%-0.5%, 0.1%-1%, or 0.5%-1% in a population of subjects without a diagnosis of the disease or disorder. In another embodiment, genetic biomarker frequencies can range higher (e.g., 0.5% to 5%) and have utility for diagnostic testing or drug development targeting the genes that harbor such variants. Genetic variants of appreciable frequency and phenotypic effect in the general population are sometimes described as goldilocks variants (e.g., see Cohen J Clin Lipidol. 2013 May-June; 7(3 Suppl):S1-5 and Price et al. Am J Hum Genet. 2010 Jun. 11; 86(6):832-8).


In some embodiments, the presence or absence of the disease or disorder in the subject can be determined with at least 50% confidence. For example, the presence or absence of the disease or disorder in the subject can be determined with at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% confidence. In some embodiments, the presence or absence of the disease or disorder in the subject can be determined with a 50%-100% confidence. For example, the presence or absence of the disease or disorder in the subject can be determined with a 60%-100%, 70%-100%, 80%-100%, 90%-100%, 50%-90%, 50%-80%, 50%-70%, 50%-60%, 60%-90%, 60%-80%, 60%-70%, 70%-90%, 70%-80%, or 80%-90%. In one embodiment, PML candidate CNVs and genes or regulatory loci associated with these CNVs can be determined or identified by comparing genetic data from a cohort of normal individuals to that of an individual or a cohort of individuals known to have, or be susceptible to PML.


In one embodiment, PML candidate CNV-subregions and genes associated with these regions can be determined or identified by comparing genetic data from a cohort of normal individuals, such as a pre-existing database of CNVs found in normal individuals termed the Normal Variation Engine (NVE), to that of a cohort of individual known to have, or be susceptible to PML.


In some embodiments, a nucleic acid sample from one individual or nucleic acid samples from a pool of 2 or more individuals without PML can serve as the reference nucleic acid sample(s) and the nucleic acid sample from an individual known to have PML or being tested to determine if they have PML can serve as the test nucleic acid sample. In one preferred embodiment, the reference and test nucleic acid samples are sex-matched and co-hybridized on the CGH array. For example, reference nucleic acid samples can be labeled with a fluorophore such as Cy5, using methods described herein, and test subject nucleic acid samples can be labeled with a different fluorophore, such as Cy3. After labeling, nucleic acid samples can be combined and can be co-hybridized to a microarray and analyzed using any of the methods described herein, such as aCGH. Arrays can then be scanned and the data can be analyzed with software. Genetic alterations, such as CNVs, can be called using any of the methods described herein. A list of the genetic alterations, such as CNVs, can be generated for one or more test subjects and/or for one or more reference subjects. Such lists of CNVs can be used to generate a master list of non-redundant CNVs and/or CNV-subregions for each type of cohort. In one embodiment, a cohort of test nucleic acid samples, such as individuals known to have or suspected to have PML, can be cohybridized with an identical sex-matched reference individual or sex-matched pool of reference individuals to generate a list of redundant or non-redundant CNVs. Such lists can be based on the presence or absence of one or more CNVs and/or CNV subregions present in individuals within the cohort. In this manner, a master list can contain a number of distinct CNVs and/or CNV-subregions, some of which are uniquely present in a single individual and some of which are present in multiple individuals.


In some embodiments, CNVs and/or CNV-subregions of interest can be obtained by annotation of each CNV and/or CNV-subregion with relevant information, such as overlap with known genes and/or exons or intergenic regulatory regions such as transcription factor binding sites. In some embodiments, CNVs and/or CNV-subregions of interest can be obtained by calculating the OR for a CNV and/or CNV-subregion according to the following formula: OR=(PML/((#individuals in PML cohort)−PML))/(NVE/((#individuals in NVE cohort)−NVE)), where: PML=number of PML individuals with a CNV-subregion of interest and NVE=number of NVE subjects with the CNV-subregion of interest. If NVE=0, it can be set to 1 to avoid dealing with infinities in cases where no CNVs are seen in the NVE. In some embodiments, a set of publicly available CNVs (e.g., the Database of Genomic Variants) can be used as the Normal cohort for comparison to the affected cohort CNVs. In another embodiment, the set of Normal cohort CNVs may comprise a private database generated by the same CNV detection method, such as array CGH, or by a plurality of CNV detection methods that include, but are not limited to, array CGH, SNP genotyping arrays, custom CGH arrays, custom genotyping arrays, exome sequencing, whole genome sequencing, targeted sequencing, FISH, q-PCR, or MLPA.


The number of individuals in any given cohort can be at least about 10, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2500, 5000, 7500, 10,000, 100,000, or more. In some embodiments, the number of individuals in any given cohort can be from 25-900, 25-800, 25-700, 25-600, 25-500, 25-400, 25-300, 25-200, 25-100, 100-1000, 100-900, 100-800, 100-700, 100-600, 100-500, 100-400, 100-300, 100-200, 200-1000, 200-900, 200-800, 200-700, 200-600, 200-500, 200-400, 200-300, 300-1000, 300-900, 300-800, 300-700, 300-600, 300-500, 300-400, 400-1000, 400-900, 400-800, 400-700, 400-600, 400-500, 500-1000, 500-900, 500-800, 500-700, 500-600, 600-1000, 600-900, 600-800, 600-700, 700-1000, 700-900, 700-800, 800-1000, 800-900, or 900-1000.


In some embodiments, a method of determining relevance or statistical significance of a genetic variant in a human subject to a disease or a condition associated with a genotype comprising screening a genome of a human subject with the disease or condition, such as by array Comparative Genomic Hybridization, sequencing, or SNP genotyping, to provide information on one or more genetic variants, such as those in Tables 1 and 2. The method can further comprise comparing, such as via a computer, information of said one or more genetic variants from the genome of said subject to a compilation of data comprising frequencies of genetic variants in at least 100 normal human subjects, such as those without the disease or condition. The method can further comprise determining a statistical significance or relevance of said one or more genetic variants from said comparison to the condition or disease or determining whether a genetic variant is present in said human subject but not present in said compilation of data from said comparison, or an algorithm can be used to call or identify significant genetic variations, such as a genetic variation whose median log 2 ratio is above or below a computed value. A computer can comprise computer executable logic that provides instructions for executing said comparison.


Different categories for CNVs of interest can be defined. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions occur within intergenic regions and are associated with an OR of at least 0.7. For example, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions occur within intergenic regions and are associated with an OR of at least 0.7, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 175, or more. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions occur within intergenic regions and are associated with an OR from about 0.7-200, 0.7-200, 0.7-90, 0.7-80, 0.7-70, 0.7-60, 0.7-50, 0.7-40, 0.7-30, 0.7-20, 0.7-10, 0.7-5, 10-200, 10-180, 10-160, 10-140, 10-120, 10-100, 10-80, 10-60, 10-40, 10-20, 20-200, 20-180, 20-160, 20-140, 20-120, 20-100, 20-80, 20-60, 20-40, 30-200, 30-180, 30-160, 30-140, 30-120, 30-100, 30-80, 30-60, 30-40, 40-200, 40-180, 40-160, 40-140, 40-120, 40-100, 40-90, 40-80, 40-70, 40-60, 40-50, 50-200, 50-180, 50-160, 50-140, 50-120, 50-100, 50-90, 50-80, 50-70, 50-60, 60-200, 60-180, 60-160, 60-140, 60-120, 60-100, 60-90, 60-80, 60-70, 70-200, 70-180, 70-160, 70-140, 70-120, 70-100, 70-90, 70-80, 80-200, 80-180, 80-160, 80-140, 80-120, 80-100, 80-90, 90-200, 90-180, 90-160, 90-140, 90-120, or 90-100.


In some embodiments, CNVs/CNV-subregions can be of interest if the CNV/CNV-subregion overlaps a known gene, and is associated with an OR of at least 1.8. For example, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions occur within intergenic regions and are associated with an OR of at least 1.8, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 175, or more. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions occur within exonic regions and are associated with an OR from about 1.8-200, 1.8-200, 1.8-90, 1.8-80, 1.8-70, 1.8-60, 1.8-50, 1.8-40, 1.8-30, 1.8-20, 1.8-10, 1.8-5, 10-200, 10-180, 10-160, 10-140, 10-120, 10-100, 10-80, 10-60, 10-40, 10-20, 20-200, 20-180, 20-160, 20-140, 20-120, 20-100, 20-80, 20-60, 20-40, 30-200, 30-180, 30-160, 30-140, 30-120, 30-100, 30-80, 30-60, 30-40, 40-200, 40-180, 40-160, 40-140, 40-120, 40-100, 40-90, 40-80, 40-70, 40-60, 40-50, 50-200, 50-180, 50-160, 50-140, 50-120, 50-100, 50-90, 50-80, 50-70, 50-60, 60-200, 60-180, 60-160, 60-140, 60-120, 60-100, 60-90, 60-80, 60-70, 70-200, 70-180, 70-160, 70-140, 70-120, 70-100, 70-90, 70-80, 80-200, 80-180, 80-160, 80-140, 80-120, 80-100, 80-90, 90-200, 90-180, 90-160, 90-140, 90-120, or 90-100.


In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 1 or more PML cases but only 0 Normal subjects. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 2 or more PML cases but only 0 or 1 Normal subjects. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 1-5 PML cases but only 0 or 1 Normal subjects. For example, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 1 PML case but only 0 or 1 Normal subjects. This can enable identification of rarer CNVs in cases with PML. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 1 PML case but only 0 or 1 Normal subjects, and are associated with an OR greater than 0.7, such as 1.8. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 2 PML cases but only 0 or 1 Normal subjects. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 3 PML cases but only 0 or 1 Normal subjects. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 4 PML cases but only 0 or 1 Normal subjects.


In some embodiments, CNVs/CNV-subregions can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is at least 0.67. For example, a CNV/CNV-subregion can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is at least 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 175, or more. In some embodiments, a CNVs/CNV-subregions can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is from about 0.7-200, 0.7-200, 0.7-90, 0.7-80, 0.7-70, 0.7-60, 0.7-50, 0.7-40, 0.7-30, 0.7-20, 0.7-10, 0.7-5, 10-200, 10-180, 10-160, 10-140, 10-120, 10-100, 10-80, 10-60, 10-40, 10-20, 20-200, 20-180, 20-160, 20-140, 20-120, 20-100, 20-80, 20-60, 20-40, 30-200, 30-180, 30-160, 30-140, 30-120, 30-100, 30-80, 30-60, 30-40, 40-200, 40-180, 40-160, 40-140, 40-120, 40-100, 40-90, 40-80, 40-70, 40-60, 40-50, 50-200, 50-180, 50-160, 50-140, 50-120, 50-100, 50-90, 50-80, 50-70, 50-60, 60-200, 60-180, 60-160, 60-140, 60-120, 60-100, 60-90, 60-80, 60-70, 70-200, 70-180, 70-160, 70-140, 70-120, 70-100, 70-90, 70-80, 80-200, 80-180, 80-160, 80-140, 80-120, 80-100, 80-90, 90-200, 90-180, 90-160, 90-140, 90-120, or 90-100.


In some embodiments, CNVs/CNV-subregions can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is at least 1.8. For example, a CNV/CNV-subregion can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is at least 1.8, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 175, or more. In some embodiments, a CNVs/CNV-subregions can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is from about 1.8-200, 1.8-200, 1.8-90, 1.8-80, 1.8-70, 1.8-60, 1.8-50, 1.8-40, 1.8-30, 1.8-20, 1.8-10, 1.8-5, 10-200, 10-180, 10-160, 10-140, 10-120, 10-100, 10-80, 10-60, 10-40, 10-20, 20-200, 20-180, 20-160, 20-140, 20-120, 20-100, 20-80, 20-60, 20-40, 30-200, 30-180, 30-160, 30-140, 30-120, 30-100, 30-80, 30-60, 30-40, 40-200, 40-180, 40-160, 40-140, 40-120, 40-100, 40-90, 40-80, 40-70, 40-60, 40-50, 50-200, 50-180, 50-160, 50-140, 50-120, 50-100, 50-90, 50-80, 50-70, 50-60, 60-200, 60-180, 60-160, 60-140, 60-120, 60-100, 60-90, 60-80, 60-70, 70-200, 70-180, 70-160, 70-140, 70-120, 70-100, 70-90, 70-80, 80-200, 80-180, 80-160, 80-140, 80-120, 80-100, 80-90, 90-200, 90-180, 90-160, 90-140, 90-120, or 90-100.


In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions do not overlap (distinct CNV/CNV-subregion), but impact the same gene (or regulatory locus) and are associated with an OR of at least 6 (Genic (distinct CNV-subregions); OR>6). For example, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions do not overlap, but impact the same gene (or regulatory locus), and are associated with an OR of at 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, or more. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions do not overlap, but impact the same gene (or regulatory locus), and are associated with an OR from about 6-100, 6-50, 6-40, 6-30, 6-20, 6-10, 6-9, 6-8, 6-7, 8-100, 8-50, 8-40, 8-30, 8-20, 8-10, 10-100, 10-50, 10-40, 10-30, 10-20, 20-100, 20-50, 20-40, 20-30, 30-100, 30-50, 30-40, 40-100, 40-50, 50-100, or 5-7. The CNV-subregion/gene can be an exonic or intronic part of the gene, or both.


In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions do not overlap a known gene (e.g., are non-genic or intergenic) and they are associated with an OR of at least 7 (Exon+ve, PML>4, NVE<2). For example, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregion does not overlap a known gene (e.g., is non-genic or intergenic) and/or non-overlapping, impact an exon, affect 2 or more PML cases but only 0 or 1 Normal subjects and are associated with an OR of at least 8, 9, 10, 11, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, or more. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, affect 2 or more PML cases but only 0 or 1 Normal subjects and are associated with an OR from about 7-100, 7-50, 7-40, 7-30, 7-20, 20-100, 20-50, 20-40, 20-30, 30-100, 30-50, 30-40, 40-100, 40-50, 50-100, or 7-11.


In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 1-5 PML cases but only 0 or 1 Normal subjects. This can enable identification of rarer CNVs in cases with PML. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 1 PML case but only 0 or 1 Normal subjects, and are associated with an OR greater than 1, such as 1.47, or from 1-2.5. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 2 PML cases but only 0 or 1 Normal subjects and are associated with an OR greater than 2.5, such as 2.95, or from 2.5-4. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 3 PML cases but only 0 or 1 Normal subjects and are associated with an OR greater than 4, such as 4.44, or from 4-5.5. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions are overlapping and/or non-overlapping, impact an exon, and they affect 4 PML cases but only 0 or 1 Normal subjects and are associated with an OR greater than 5.5, such as 5.92, or from 5.5-6.8.


In some embodiments, CNVs/CNV-subregions can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is at least 6. For example, a CNV/CNV-subregion can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is at least 7, 8, 9, 10, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, or more. In some embodiments, a CNVs/CNV-subregions can be of interest if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNVs/CNV-subregions) is from about 6-100, 6-50, 6-40, 6-30, 6-20, 6-10, 6-9, 6-8, 6-7, 8-100, 8-50. 8-40, 8-30, 8-20, 8-10, 10-100, 10-50, 10-40, 10-30, 10-20, 20-100, 20-50, 20-40, 20-30, 30-100, 30-50, 30-40, 40-100, 40-50, 50-100, or 5-7.


In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions impact an intron and they affect 5 or more PML cases but only 0 or 1 Normal subjects and they are associated with an OR of at least 7 (Intron+ve, PML>4, Normals<2). For example, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions impact an intron and they affect 5 or more PML cases but only 0 or 1 Normal subjects and they are associated with an OR of at least 8, 9, 10, 11, 12, 14, 16, 18, 20, 25, 30, 35, 40, 45, 50, or more. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions impact an intron and they affect 5 or more PML cases but only 0 or 1 Normal subjects and they are associated with an OR from about 7-100, 7-50, 7-40, 7-30, 7-20, 20-100, 20-50, 20-40, 20-30, 30-100, 30-50, 30-40, 40-100, 40-50, 50-100, or 7-11. CNVs/CNV-subregions impacting introns can be pathogenic (e.g., such variants can result in alternatively spliced mRNAs or loss of a microRNA binding site, which may deleteriously impact the resulting protein's structure or expression level).


In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions occur within intergenic regions and are associated with an OR of greater than 30 (High OR intergenic (OR>30)). For example, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions occur within intergenic regions and are associated with an OR of greater than 31, 32, 33, 34, 35, 40, 45, 50, 66, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more. In some embodiments, CNVs/CNV-subregions can be of interest if the CNVs/CNV-subregions impact occur within intergenic regions and are associated with an OR from about 30-100, 30-90, 30-80, 30-70, 30-60, 30-50, 30-40, 40-100, 40-90, 40-80, 40-70, 40-60, 40-50, 50-100, 50-90, 50-80, 50-70, 50-60, 60-100, 60-90, 60-80, 60-70, 70-100, 70-90, 70-80, 80-100, 80-90, or 90-100.


In some embodiments, a CNV/CNV-subregion can be of interest if the CNV/CNV-subregion overlaps a known gene, and is associated with an OR of at least 10. In some embodiments, a CNV/CNV-subregion can be of interest if the CNV/CNV-subregion overlaps a known gene, is associated with an OR of at least 6, and if the OR associated with the sum of PML cases and the sum of NVE subjects affecting the same gene (including distinct CNV-subregions) is at least 6.


Methods of Treatment


One embodiment of the present disclosure provides methods, pharmaceutical compositions, and kits for the treatment of a condition in animal subjects. The condition can be HIV/AIDS, cancer, or an autoimmune disease. In some embodiments, the condition can be PML. For example, the condition can be multiple sclerosis. In some embodiments, the methods comprise administering one or more immunosuppressive medications. In some embodiments, the pharmaceutical compositions and kits comprise one or more immunosuppressive medications. The one or more immunosuppressive medications can be adalimumab (e.g., Humira), alemtuzumab (e.g., Lemtrada), alentuzumab (e.g., Campath), azathioprine (e.g., Imuran), belimumab (e.g., Benlysta), bevacizumab (e.g., Avastatin), bortezomib (e.g., Velcade), eculizumab (e.g., Soliris), leflunomide, brentuximab vedotin (e.g., Adcetris), cetuximab (e.g., Erbitux), cyclophosphamid, dimethyl fumarate (e.g., Tecfidera), efalizumab (e.g., Raptiva), fingolimod (e.g., Gilenya), fludarabine (e.g., Fludara), fumaric acid, imatinib (e.g., Gleevec, Glivec), infliximab (e.g., Remicade), methotrexate (e.g., Trexall, Rheumatrex), mycophenolate mofetil (e.g., Cellcept), natalizumab (e.g., Tysabri), rituximab (e.g., Rituxin), daclizumab (e.g., Zinbryta), vedolizumab (Entyvio), ruxolitinib (e.g., Jakafi, Jakavi), ocrelizumab (e.g., Ocrevus), or any combinations thereof. The term “animal subject” as used herein includes humans as well as other mammals. The term “treating” as used herein includes achieving a therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying viral infection (e.g., HIV), cancer, or autoimmune disease.


In some embodiments, a subject can be currently treated with an antiretroviral medication. In some embodiments, a subject can be previously treated with an antiretroviral medication. In some embodiments, a subject can be not yet treated with an antiretroviral medication. The antiretroviral medication can include but not limited to Nucleoside Reverse Transcriptase Inhibitors (NRTIs), Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTIs), Protease Inhibitors (PIs), Fusion Inhibitors, Entry Inhibitors, Integrase Inhibitors, Pharmacokinetic Enhancers, and Combination HIV Medicines. In some cases, the Nucleoside Reverse Transcriptase Inhibitors can include but not limited to abacavir, didanosine, emtricitabine, lamivudine, stavudine, tenofovir disoproxil fumarate, and zidovudine. In some cases, the Non-Nucleoside Reverse Transcriptase Inhibitors can include but not limited to efavirenz, etravirine, nevirapine, and rilpivirine. In some cases, the Protease Inhibitors can include but not limited to atazanavir, darunavir, fosamprenavir, indinavir, nelfinavir, ritonavir, saquinavir, and tipranavir. In some cases, the Fusion Inhibitors can include but not limited to enfuvirtide. In some cases, the Entry Inhibitors can include but not limited to maraviroc. In some cases, the Integrase Inhibitors can include but not limited to dolutegravir, elvitegravir, and raltegravir. In some cases, the Pharmacokinetic Enhancers can include but not limited to cobicistat. In some cases, the Combination HIV Medicines can include but not limited to abacavir and lamivudine, abacavir, dolutegravir, and lamivudine, abacavir, lamivudine, and zidovudine, atazanavir and cobicistat, darunavir and cobicistat, efavirenz, emtricitabine, and tenofovir disoproxil fumarate, elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide fumarate, elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate, emtricitabine, rilpivirine, and tenofovir alafenamide, emtricitabine, rilpivirine, and tenofovir disoproxil fumarate, emtricitabine and tenofovir alafenamide, emtricitabine and tenofovir disoproxil fumarate, lamivudine and zidovudine, lopinavir and ritonavir, and any combination of antiretroviral medications listed above.


In some embodiments, such as when a subject is identified as having at least one of the genetic variants described herein, an agent targeting the JC Virus can be administered to the subject. In some embodiments, a medication can be administered to a subject that prevents PML from developing, or it can reduce, lessen, shorten and/or otherwise ameliorate the progression of PML, or symptoms that develop. The pharmaceutical composition can modulate or target JC Virus. In some embodiments, a subject identified as having PML can be administered an agent that reduces a viral load in the subject. In some embodiments, an immunosuppressive agent can be administered prior to, or in conjunction with, an agent that reduces a viral load in the subject. In some embodiments, a subject identified as having a risk of developing PML can be administered an agent that prevents an increase in a viral load in the subject. In some embodiments, a subject identified as having a high risk of developing PML can be administered an agent that prevents an increase in a viral load in the subject. In some embodiments, an immunosuppressive agent can be administered prior to, or in conjunction with, an agent that prevents an increase in a viral load in the subject. The agent that reduces a viral load in the subject or that prevents an increase in a viral load in the subject can be, for example, an agent that targets JC Virus. Exemplary agents include antibodies, such as broadly neutralizing JCV antibodies. For example, an agent can be a broadly neutralizing human monoclonal JC polyomavirus VP-1 specific antibody (See, e.g., Jelcic et al., Science Translational Medicine, Vol. 7, Issue 306, pp. 306ra150 (2015) and Ray et al., Science Translational Medicine, Vol. 7, Issue 306, pp 306ra151 (2015)). Additional exemplary agents include antiretroviral agents, cidofovir, hexadecyloxypropyl-cidofovir (a lipid-ester derivative), cytarabine (cytosine arabinoside), agents that block the 5HT2a receptor (e.g., olanzapine, zisprasidone, mirtazapine, cyproheptadine, and risperidone), topoisomerase inhibitors (e.g., topotecan), and mefloquine.


In some embodiments, a pharmaceutical composition of the disclosure can be administered to a subject at risk of developing PML, or to a subject reporting one or more of the physiological symptoms of PML, even though a screening of the condition cannot have been made. In some embodiments, a pharmaceutical composition of the disclosure can be administered to a subject not identified as having a risk of developing PML, or to a subject not identified as having one or more of the physiological symptoms of PML, even though a screening of the condition cannot have been made.


The present disclosure also includes kits that can be used to treat a condition in animal subjects. These kits comprise one or more immunosuppressive medications and in some embodiments instructions teaching the use of the kit according to the various methods and approaches described herein. Such kits can also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages (or risks and/or disadvantages) of the agent. Such information can be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like.


In some aspects a host cell can be used for testing or administering therapeutics. In some embodiments, a host cell can comprise a nucleic acid comprising expression control sequences operably-linked to a coding region. The host cell can be natural or non-natural. The non-natural host used in aspects of the method can be any cell capable of expressing a nucleic acid of the disclosure including, bacterial cells, fungal cells, insect cells, mammalian cells and plant cells. In some aspects the natural host is a mammalian tissue cell and the non-natural host is a different mammalian tissue cell. Other aspects of the method include a natural host that is a first cell normally residing in a first mammalian species and the non-natural host is a second cell normally residing in a second mammalian species. In another alternative aspect, the method uses a first cell and the second cell that are from the same tissue type. In those aspects of the method where the coding region encodes a mammalian polypeptide, the mammalian polypeptide may be a hormone. In other aspects the coding region may encode a neuropeptide, an antibody, an antimetabolite, or a polypeptide or nucleotide therapeutic.


Expression control sequences can be those nucleotide sequences, both 5′ and 3′ to a coding region, that are required for the transcription and translation of the coding region in a host organism. Regulatory sequences include a promoter, ribosome binding site, optional inducible elements and sequence elements required for efficient 3′ processing, including polyadenylation. When the structural gene has been isolated from genomic DNA, the regulatory sequences also include those intronic sequences required for splicing of the introns as part of mRNA formation in the target host.


Formulations, Routes of Administration, and Effective Doses


Yet another aspect of the present disclosure relates to formulations, routes of administration and effective doses for pharmaceutical compositions comprising an agent or combination of agents of the instant disclosure. Such pharmaceutical compositions can be used to treat a condition (e.g., multiple sclerosis) as described above.


Compounds of the disclosure can be administered as pharmaceutical formulations including those suitable for oral (including buccal and sub-lingual), rectal, nasal, topical, transdermal patch, pulmonary, vaginal, suppository, or parenteral (including intramuscular, intraarterial, intrathecal, intradermal, intraperitoneal, subcutaneous and intravenous) administration or in a form suitable for administration by aerosolization, inhalation or insufflation. General information on drug delivery systems can be found in Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems (Lippencott Williams & Wilkins, Baltimore Md. (1999).


In various embodiments, the pharmaceutical composition includes carriers and excipients (including but not limited to buffers, carbohydrates, mannitol, polypeptides, amino acids, antioxidants, bacteriostats, chelating agents, suspending agents, thickening agents and/or preservatives), water, oils including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like, saline solutions, aqueous dextrose and glycerol solutions, flavoring agents, coloring agents, detackifiers and other acceptable additives, adjuvants, or binders, other pharmaceutically acceptable auxiliary substances to approximate physiological conditions, such as pH buffering agents, tonicity adjusting agents, emulsifying agents, wetting agents and the like. Examples of excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. In some embodiments, the pharmaceutical preparation is substantially free of preservatives. In other embodiments, the pharmaceutical preparation can contain at least one preservative. General methodology on pharmaceutical dosage forms is found in Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems (Lippencott, Williams, & Wilkins, Baltimore Md. (1999)). It can be recognized that, while any suitable carrier known to those of ordinary skill in the art can be employed to administer the compositions of this disclosure, the type of carrier can vary depending on the mode of administration.


Compounds can also be encapsulated within liposomes using well-known technology. Biodegradable microspheres can also be employed as carriers for the pharmaceutical compositions of this disclosure. Suitable biodegradable microspheres are disclosed, for example, in U.S. Pat. Nos. 4,897,268, 5,075,109, 5,928,647, 5,811,128, 5,820,883, 5,853,763, 5,814,344 and 5,942,252.


The compound can be administered in liposomes or microspheres (or microparticles). Methods for preparing liposomes and microspheres for administration to a subject are well known to those of skill in the art. U.S. Pat. No. 4,789,734, the contents of which are hereby incorporated by reference, describes methods for encapsulating biological materials in liposomes. Essentially, the material is dissolved in an aqueous solution, the appropriate phospholipids and lipids added, and along with surfactants if required, and the material dialyzed or sonicated, as necessary. A review of known methods is provided by G. Gregoriadis, Chapter 14, “Liposomes,” Drug Carriers in Biology and Medicine, pp. 2.sup.87-341 (Academic Press, 1979).


Microspheres formed of polymers or polypeptides are well known to those skilled in the art, and can be tailored for passage through the gastrointestinal tract directly into the blood stream. Alternatively, the compound can be incorporated and the microspheres, or composite of microspheres, implanted for slow release over a period of time ranging from days to months. See, for example, U.S. Pat. Nos. 4,906,474, 4,925,673 and 3,625,214, and Jein, TIPS 19:155-157 (1998), the contents of which are hereby incorporated by reference.


The concentration of drug can be adjusted, the pH of the solution buffered and the isotonicity adjusted to be compatible with intravenous injection, as is well known in the art.


The compounds of the disclosure can be formulated as a sterile solution or suspension, in suitable vehicles, well known in the art. The pharmaceutical compositions can be sterilized by conventional, well-known sterilization techniques, or can be sterile filtered. The resulting aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration. Suitable formulations and additional carriers are described in Remington “The Science and Practice of Pharmacy” (20th Ed., Lippincott Williams & Wilkins, Baltimore MD), the teachings of which are incorporated by reference in their entirety herein.


The agents or their pharmaceutically acceptable salts can be provided alone or in combination with one or more other agents or with one or more other forms. For example, a formulation can comprise one or more agents in particular proportions, depending on the relative potencies of each agent and the intended indication. For example, in compositions for targeting two different host targets, and where potencies are similar, about a 1:1 ratio of agents can be used. The two forms can be formulated together, in the same dosage unit e.g., in one cream, suppository, tablet, capsule, aerosol spray, or packet of powder to be dissolved in a beverage; or each form can be formulated in a separate unit, e.g., two creams, two suppositories, two tablets, two capsules, a tablet and a liquid for dissolving the tablet, two aerosol sprays, or a packet of powder and a liquid for dissolving the powder, etc.


The term “pharmaceutically acceptable salt” means those salts which retain the biological effectiveness and properties of the agents used in the present disclosure, and which are not biologically or otherwise undesirable.


Typical salts are those of the inorganic ions, such as, for example, sodium, potassium, calcium, magnesium ions, and the like. Such salts include salts with inorganic or organic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, nitric acid, sulfuric acid, methanesulfonic acid, p toluenesulfonic acid, acetic acid, fumaric acid, succinic acid, lactic acid, mandelic acid, malic acid, citric acid, tartaric acid or maleic acid. In addition, if the agent(s) contain a carboxy group or other acidic group, it can be converted into a pharmaceutically acceptable addition salt with inorganic or organic bases. Examples of suitable bases include sodium hydroxide, potassium hydroxide, ammonia, cyclohexylamine, dicyclohexyl-amine, ethanolamine, diethanolamine, triethanolamine, and the like.


A pharmaceutically acceptable ester or amide refers to those which retain biological effectiveness and properties of the agents used in the present disclosure, and which are not biologically or otherwise undesirable. Typical esters include ethyl, methyl, isobutyl, ethylene glycol, and the like. Typical amides include unsubstituted amides, alkyl amides, dialkyl amides, and the like.


In some embodiments, an agent can be administered in combination with one or more other compounds, forms, and/or agents, e.g., as described above. Pharmaceutical compositions with one or more other active agents can be formulated to comprise certain molar ratios. For example, molar ratios of about 99:1 to about 1:99 of a first active agent to the other active agent can be used. In some subset of the embodiments, the range of molar ratios of a first active agent: other active agents are selected from about 80:20 to about 20:80; about 75:25 to about 25:75, about 70:30 to about 30:70, about 66:33 to about 33:66, about 60:40 to about 40:60; about 50:50; and about 90:10 to about 10:90. The molar ratio of a first active:other active agents can be about 1:9, and in some embodiments can be about 1:1. The two agents, forms and/or compounds can be formulated together, in the same dosage unit e.g., in one cream, suppository, tablet, capsule, or packet of powder to be dissolved in a beverage; or each agent, form, and/or compound can be formulated in separate units, e.g., two creams, suppositories, tablets, two capsules, a tablet and a liquid for dissolving the tablet, an aerosol spray a packet of powder and a liquid for dissolving the powder, etc.


If necessary or desirable, the agents and/or combinations of agents can be administered with still other agents. The choice of agents that can be co-administered with the agents and/or combinations of agents of the instant disclosure can depend, at least in part, on the condition being treated. Agents of particular use in the formulations of the present disclosure include, for example, any agent having a therapeutic effect for a viral infection, including, e.g., drugs used to treat inflammatory conditions. For example, in treatments for influenza, in some embodiments formulations of the instant disclosure can additionally contain one or more conventional anti-inflammatory drugs, such as an NSAID, e.g., ibuprofen, naproxen, acetaminophen, ketoprofen, or aspirin. In some alternative embodiments for the treatment of influenza formulations of the instant disclosure can additionally contain one or more conventional influenza antiviral agents, such as amantadine, rimantadine, zanamivir, and oseltamivir. In treatments for retroviral infections, such as HIV, formulations of the instant disclosure can additionally contain one or more conventional antiviral drug, such as protease inhibitors (lopinavir/ritonavir {Kaletra}, indinavir {Crixivan}, ritonavir {Norvir}, nelfinavir {Viracept}, saquinavir hard gel capsules {Invirase}, atazanavir {Reyataz}, amprenavir {Agenerase}, fosamprenavir {Telzir}, tipranavir{Aptivus}), reverse transcriptase inhibitors, including non-Nucleoside and Nucleoside/nucleotide inhibitors (AZT {zidovudine, Retrovir}, ddI {didanosine, Videx}, 3TC {lamivudine, Epivir}, d4T {stavudine, Zerit}, abacavir {Ziagen}, FTC {emtricitabine, Emtriva}, tenofovir {Viread}, efavirenz {Sustiva} and nevirapine {Viramune}), fusion inhibitors T20 {enfuvirtide, Fuzeon}, integrase inhibitors (MK-0518 and GS-9137), and maturation inhibitors (PA-457 {Bevirimat}). As another example, formulations can additionally contain one or more supplements, such as vitamin C, E or other anti-oxidants.


The agent(s) (or pharmaceutically acceptable salts, esters or amides thereof) can be administered per se or in the form of a pharmaceutical composition wherein the active agent(s) is in an admixture or mixture with one or more pharmaceutically acceptable carriers. A pharmaceutical composition, as used herein, can be any composition prepared for administration to a subject. Pharmaceutical compositions for use in accordance with the present disclosure can be formulated in conventional manner using one or more physiologically acceptable carriers, comprising excipients, diluents, and/or auxiliaries, e.g., which facilitate processing of the active agents into preparations that can be administered. Proper formulation can depend at least in part upon the route of administration chosen. The agent(s) useful in the present disclosure, or pharmaceutically acceptable salts, esters, or amides thereof, can be delivered to a subject using a number of routes or modes of administration, including oral, buccal, topical, rectal, transdermal, transmucosal, subcutaneous, intravenous, and intramuscular applications, as well as by inhalation.


For oral administration, the agents can be formulated readily by combining the active agent(s) with pharmaceutically acceptable carriers well known in the art. Such carriers enable the agents of the disclosure to be formulated as tablets, including chewable tablets, pills, dragees, capsules, lozenges, hard candy, liquids, gels, syrups, slurries, powders, suspensions, elixirs, wafers, and the like, for oral ingestion by a subject to be treated. Such formulations can comprise pharmaceutically acceptable carriers including solid diluents or fillers, sterile aqueous media and various non-toxic organic solvents. A solid carrier can be one or more substances which can also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. In powders, the carrier generally is a finely divided solid which is a mixture with the finely divided active component. In tablets, the active component generally is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted in the shape and size desired. The powders and tablets preferably contain from about one (1) to about seventy (70) percent of the active compound. Suitable carriers include but are not limited to magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like. Generally, the agents of the disclosure can be included at concentration levels ranging from about 0.5%, about 5%, about 10%, about 20%, or about 30% to about 50%, about 60%, about 70%, about 80% or about 90% by weight of the total composition of oral dosage forms, in an amount sufficient to provide a desired unit of dosage.


Aqueous suspensions for oral use can contain agent(s) of this disclosure with pharmaceutically acceptable excipients, such as a suspending agent (e.g., methyl cellulose), a wetting agent (e.g., lecithin, lysolecithin and/or a long-chain fatty alcohol), as well as coloring agents, preservatives, flavoring agents, and the like.


In some embodiments, oils or non-aqueous solvents can be used to bring the agents into solution, due to, for example, the presence of large lipophilic moieties. Alternatively, emulsions, suspensions, or other preparations, for example, liposomal preparations, can be used. With respect to liposomal preparations, any known methods for preparing liposomes for treatment of a condition can be used. See, for example, Bangham et al., J. Mol. Biol. 23: 238-252 (1965) and Szoka et al., Proc. Natl Acad. Sci. USA 75: 4194-4198 (1978), incorporated herein by reference. Ligands can also be attached to the liposomes to direct these compositions to particular sites of action. Agents of this disclosure can also be integrated into foodstuffs, e.g., cream cheese, butter, salad dressing, or ice cream to facilitate solubilization, administration, and/or compliance in certain subject populations.


Pharmaceutical preparations for oral use can be obtained as a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; flavoring elements, cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone (PVP). If desired, disintegrating agents can be added, such as the cross linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. The agents can also be formulated as a sustained release preparation.


Dragee cores can be provided with suitable coatings. For this purpose, concentrated sugar solutions can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active agents.


Pharmaceutical preparations that can be used orally include push fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active agents can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in dosages suitable for administration.


Other forms suitable for oral administration include liquid form preparations including emulsions, syrups, elixirs, aqueous solutions, aqueous suspensions, or solid form preparations which are intended to be converted shortly before use to liquid form preparations. Emulsions can be prepared in solutions, for example, in aqueous propylene glycol solutions or can contain emulsifying agents, for example, such as lecithin, sorbitan monooleate, or acacia. Aqueous solutions can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening agents. Aqueous suspensions can be prepared by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well known suspending agents. Suitable fillers or carriers with which the compositions can be administered include agar, alcohol, fats, lactose, starch, cellulose derivatives, polysaccharides, polyvinylpyrrolidone, silica, sterile saline and the like, or mixtures thereof used in suitable amounts. Solid form preparations include solutions, suspensions, and emulsions, and can contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.


A syrup or suspension can be made by adding the active compound to a concentrated, aqueous solution of a sugar, e.g., sucrose, to which can also be added any accessory ingredients. Such accessory ingredients can include flavoring, an agent to retard crystallization of the sugar or an agent to increase the solubility of any other ingredient, e.g., as a polyhydric alcohol, for example, glycerol or sorbitol.


When formulating compounds of the disclosure for oral administration, it can be desirable to utilize gastroretentive formulations to enhance absorption from the gastrointestinal (GI) tract. A formulation which is retained in the stomach for several hours can release compounds of the disclosure slowly and provide a sustained release that can be preferred in some embodiments of the disclosure. Disclosure of such gastro-retentive formulations are found in Klausner E. A., et al., Pharm. Res. 20, 1466-73 (2003); Hoffman, A. et al., Int. J. Pharm. 11, 141-53 (2004), Streubel, A., et al. Expert Opin. Drug Deliver. 3, 217-3, and Chavanpatil, M. D. et al., Int. J. Pharm. (2006). Expandable, floating and bioadhesive techniques can be utilized to maximize absorption of the compounds of the disclosure.


The compounds of the disclosure can be formulated for parenteral administration (e.g., by injection, for example, bolus injection or continuous infusion) and can be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, for example, solutions in aqueous polyethylene glycol.


For injectable formulations, the vehicle can be chosen from those known in art to be suitable, including aqueous solutions or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles. The formulation can also comprise polymer compositions which are biocompatible, biodegradable, such as poly(lactic-co-glycolic)acid. These materials can be made into micro or nanospheres, loaded with drug and further coated or derivatized to provide superior sustained release performance. Vehicles suitable for periocular or intraocular injection include, for example, suspensions of therapeutic agent in injection grade water, liposomes and vehicles suitable for lipophilic substances. Other vehicles for periocular or intraocular injection are well known in the art.


In some embodiments, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition can also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.


When administration is by injection, the active compound can be formulated in aqueous solutions, specifically in physiologically compatible buffers such as Hanks solution, Ringer's solution, or physiological saline buffer. The solution can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active compound can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. In some embodiments, the pharmaceutical composition does not comprise an adjuvant or any other substance added to enhance the immune response stimulated by the peptide. In some embodiments, the pharmaceutical composition comprises a substance that inhibits an immune response to the peptide. Methods of formulation are known in the art, for example, as disclosed in Remington's Pharmaceutical Sciences, latest edition, Mack Publishing Co., Easton P.


In addition to the formulations described previously, the agents can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation or transcutaneous delivery (for example, subcutaneously or intramuscularly), intramuscular injection or use of a transdermal patch. Thus, for example, the agents can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.


In some embodiments, pharmaceutical compositions comprising one or more agents of the present disclosure exert local and regional effects when administered topically or injected at or near particular sites of infection. Direct topical application, e.g., of a viscous liquid, solution, suspension, dimethylsulfoxide (DMSO)-based solutions, liposomal formulations, gel, jelly, cream, lotion, ointment, suppository, foam, or aerosol spray, can be used for local administration, to produce for example, local and/or regional effects. Pharmaceutically appropriate vehicles for such formulation include, for example, lower aliphatic alcohols, polyglycols (e.g., glycerol or polyethylene glycol), esters of fatty acids, oils, fats, silicones, and the like. Such preparations can also include preservatives (e.g., p-hydroxybenzoic acid esters) and/or antioxidants (e.g., ascorbic acid and tocopherol). See also Dermatological Formulations: Percutaneous absorption, Barry (Ed.), Marcel Dekker Incl, 1983.


Pharmaceutical compositions of the present disclosure can contain a cosmetically or dermatologically acceptable carrier. Such carriers are compatible with skin, nails, mucous membranes, tissues and/or hair, and can include any conventionally used cosmetic or dermatological carrier meeting these requirements. Such carriers can be readily selected by one of ordinary skill in the art. In formulating skin ointments, an agent or combination of agents of the instant disclosure can be formulated in an oleaginous hydrocarbon base, an anhydrous absorption base, a water-in-oil absorption base, an oil-in-water water-removable base and/or a water-soluble base. Examples of such carriers and excipients include, but are not limited to, humectants (e.g., urea), glycols (e.g., propylene glycol), alcohols (e.g., ethanol), fatty acids (e.g., oleic acid), surfactants (e.g., isopropyl myristate and sodium lauryl sulfate), pyrrolidones, glycerol monolaurate, sulfoxides, terpenes (e.g., menthol), amines, amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.


Ointments and creams can, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions can be formulated with an aqueous or oily base and can in general also containing one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches can be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.


Lubricants which can be used to form pharmaceutical compositions and dosage forms of the disclosure include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, or mixtures thereof. Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, or mixtures thereof. A lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical composition.


The compositions according to the present disclosure can be in any form suitable for topical application, including aqueous, aqueous-alcoholic or oily solutions, lotion or serum dispersions, aqueous, anhydrous or oily gels, emulsions obtained by dispersion of a fatty phase in an aqueous phase (0/W or oil in water) or, conversely, (W/O or water in oil), microemulsions or alternatively microcapsules, microparticles or lipid vesicle dispersions of ionic and/or nonionic type. These compositions can be prepared according to conventional methods. Other than the agents of the disclosure, the amounts of the various constituents of the compositions according to the disclosure are those conventionally used in the art. These compositions in particular constitute protection, treatment or care creams, milks, lotions, gels or foams for the face, for the hands, for the body and/or for the mucous membranes, or for cleansing the skin. The compositions can also consist of solid preparations constituting soaps or cleansing bars.


Compositions of the present disclosure can also contain adjuvants common to the cosmetic and dermatological fields, such as hydrophilic or lipophilic gelling agents, hydrophilic or lipophilic active agents, preserving agents, antioxidants, solvents, fragrances, fillers, sunscreens, odor-absorbers and dyestuffs. The amounts of these various adjuvants are those conventionally used in the fields considered and, for example, are from about 0.01% to about 20% of the total weight of the composition. Depending on their nature, these adjuvants can be introduced into the fatty phase, into the aqueous phase and/or into the lipid vesicles.


In some embodiments, ocular viral infections can be effectively treated with ophthalmic solutions, suspensions, ointments or inserts comprising an agent or combination of agents of the present disclosure. Eye drops can be prepared by dissolving the active ingredient in a sterile aqueous solution such as physiological saline, buffering solution, etc., or by combining powder compositions to be dissolved before use. Other vehicles can be chosen, as is known in the art, including but not limited to: balance salt solution, saline solution, water soluble polyethers such as polyethyene glycol, polyvinyls, such as polyvinyl alcohol and povidone, cellulose derivatives such as methylcellulose and hydroxypropyl methylcellulose, petroleum derivatives such as mineral oil and white petrolatum, animal fats such as lanolin, polymers of acrylic acid such as carboxypolymethylene gel, vegetable fats such as peanut oil and polysaccharides such as dextrans, and glycosaminoglycans such as sodium hyaluronate. If desired, additives ordinarily used in the eye drops can be added. Such additives include isotonizing agents (e.g., sodium chloride, etc.), buffer agent (e.g., boric acid, sodium monohydrogen phosphate, sodium dihydrogen phosphate, etc.), preservatives (e.g., benzalkonium chloride, benzethonium chloride, chlorobutanol, etc.), thickeners (e.g., saccharide such as lactose, mannitol, maltose, etc.; e.g., hyaluronic acid or its salt such as sodium hyaluronate, potassium hyaluronate, etc.; e.g., mucopolysaccharide such as chondroitin sulfate, etc.; e.g., sodium polyacrylate, carboxyvinyl polymer, crosslinked polyacrylate, polyvinyl alcohol, polyvinyl pyrrolidone, methyl cellulose, hydroxy propyl methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose, hydroxy propyl cellulose or other agents known to those skilled in the art).


The solubility of the components of the present compositions can be enhanced by a surfactant or other appropriate co-solvent in the composition. Such cosolvents include polysorbate 20, 60, and 80, Pluronic F68, F-84 and P-103, cyclodextrin, or other agents known to those skilled in the art. Such cosolvents can be employed at a level of from about 0.01% to 2% by weight.


The compositions of the disclosure can be packaged in multidose form. Preservatives can be preferred to prevent microbial contamination during use. Suitable preservatives include: benzalkonium chloride, thimerosal, chlorobutanol, methyl paraben, propyl paraben, phenylethyl alcohol, edetate disodium, sorbic acid, Onamer M, or other agents known to those skilled in the art. In the prior art ophthalmic products, such preservatives can be employed at a level of from 0.004% to 0.02%. In the compositions of the present application the preservative, preferably benzalkonium chloride, can be employed at a level of from 0.001% to less than 0.01%, e.g., from 0.001% to 0.008%, preferably about 0.005% by weight. It has been found that a concentration of benzalkonium chloride of 0.005% can be sufficient to preserve the compositions of the present disclosure from microbial attack.


In some embodiments, the agents of the present disclosure are delivered in soluble rather than suspension form, which allows for more rapid and quantitative absorption to the sites of action. In general, formulations such as jellies, creams, lotions, suppositories and ointments can provide an area with more extended exposure to the agents of the present disclosure, while formulations in solution, e.g., sprays, provide more immediate, short-term exposure.


In some embodiments relating to topical/local application, the pharmaceutical compositions can include one or more penetration enhancers. For example, the formulations can comprise suitable solid or gel phase carriers or excipients that increase penetration or help delivery of agents or combinations of agents of the disclosure across a permeability barrier, e.g., the skin. Many of these penetration-enhancing compounds are known in the art of topical formulation, and include, e.g., water, alcohols (e.g., terpenes like methanol, ethanol, 2-propanol), sulfoxides (e.g., dimethyl sulfoxide, decylmethyl sulfoxide, tetradecylmethyl sulfoxide), pyrrolidones (e.g., 2-pyrrolidone, N-methyl-2-pyrrolidone, N-(2-hydroxyethyl)pyrrolidone), laurocapram, acetone, dimethylacetamide, dimethylformamide, tetrahydrofurfuryl alcohol, L-α-amino acids, anionic, cationic, amphoteric or nonionic surfactants (e.g., isopropyl myristate and sodium lauryl sulfate), fatty acids, fatty alcohols (e.g., oleic acid), amines, amides, clofibric acid amides, hexamethylene lauramide, proteolytic enzymes, α-bisabolol, d-limonene, urea and N,N-diethyl-m-toluamide, and the like. Additional examples include humectants (e.g., urea), glycols (e.g., propylene glycol and polyethylene glycol), glycerol monolaurate, alkanes, alkanols, ORGELASE, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and/or other polymers. In some embodiments, the pharmaceutical compositions can include one or more such penetration enhancers.


In some embodiments, the pharmaceutical compositions for local/topical application can include one or more antimicrobial preservatives such as quaternary ammonium compounds, organic mercurials, p-hydroxy benzoates, aromatic alcohols, chlorobutanol, and the like.


In some embodiments, the pharmaceutical compositions can be orally- or rectally delivered solutions, suspensions, ointments, enemas and/or suppositories comprising an agent or combination of agents of the present disclosure.


In some embodiments, the pharmaceutical compositions can be aerosol solutions, suspensions or dry powders comprising an agent or combination of agents of the present disclosure. The aerosol can be administered through the respiratory system or nasal passages. For example, one skilled in the art can recognize that a composition of the present disclosure can be suspended or dissolved in an appropriate carrier, e.g., a pharmaceutically acceptable propellant, and administered directly into the lungs using a nasal spray or inhalant. For example, an aerosol formulation comprising an agent can be dissolved, suspended or emulsified in a propellant or a mixture of solvent and propellant, e.g., for administration as a nasal spray or inhalant Aerosol formulations can contain any acceptable propellant under pressure, such as a cosmetically or dermatologically or pharmaceutically acceptable propellant, as conventionally used in the art.


An aerosol formulation for nasal administration is generally an aqueous solution designed to be administered to the nasal passages in drops or sprays. Nasal solutions can be similar to nasal secretions in that they are generally isotonic and slightly buffered to maintain a pH of about 5.5 to about 6.5, although pH values outside of this range can additionally be used. Antimicrobial agents or preservatives can also be included in the formulation.


An aerosol formulation for inhalations and inhalants can be designed so that the agent or combination of agents of the present disclosure is carried into the respiratory tree of the subject when administered by the nasal or oral respiratory route. Inhalation solutions can be administered, for example, by a nebulizer. Inhalations or insufflations, comprising finely powdered or liquid drugs, can be delivered to the respiratory system as a pharmaceutical aerosol of a solution or suspension of the agent or combination of agents in a propellant, e.g., to aid in disbursement. Propellants can be liquefied gases, including halocarbons, for example, fluorocarbons such as fluorinated chlorinated hydrocarbons, hydrochlorofluorocarbons, and hydrochlorocarbons, as well as hydrocarbons and hydrocarbon ethers.


Halocarbon propellants useful in the present disclosure include fluorocarbon propellants in which all hydrogens are replaced with fluorine, chlorofluorocarbon propellants in which all hydrogens are replaced with chlorine and at least one fluorine, hydrogen-containing fluorocarbon propellants, and hydrogen-containing chlorofluorocarbon propellants. Halocarbon propellants are described in Johnson, U.S. Pat. No. 5,376,359; Byron et al., U.S. Pat. No. 5,190,029; and Purewal et al., U.S. Pat. No. 5,776,434. Hydrocarbon propellants useful in the disclosure include, for example, propane, isobutane, n-butane, pentane, isopentane and neopentane. A blend of hydrocarbons can also be used as a propellant. Ether propellants include, for example, dimethyl ether as well as the ethers. An aerosol formulation of the disclosure can also comprise more than one propellant. For example, the aerosol formulation can comprise more than one propellant from the same class, such as two or more fluorocarbons; or more than one, more than two, more than three propellants from different classes, such as a fluorohydrocarbon and a hydrocarbon. Pharmaceutical compositions of the present disclosure can also be dispensed with a compressed gas, e.g., an inert gas such as carbon dioxide, nitrous oxide or nitrogen.


Aerosol formulations can also include other components, for example, ethanol, isopropanol, propylene glycol, as well as surfactants or other components such as oils and detergents. These components can serve to stabilize the formulation and/or lubricate valve components.


The aerosol formulation can be packaged under pressure and can be formulated as an aerosol using solutions, suspensions, emulsions, powders and semisolid preparations. For example, a solution aerosol formulation can comprise a solution of an agent of the disclosure in (substantially) pure propellant or as a mixture of propellant and solvent. The solvent can be used to dissolve the agent and/or retard the evaporation of the propellant. Solvents useful in the disclosure include, for example, water, ethanol and glycols. Any combination of suitable solvents can be use, optionally combined with preservatives, antioxidants, and/or other aerosol components.


An aerosol formulation can also be a dispersion or suspension. A suspension aerosol formulation can comprise a suspension of an agent or combination of agents of the instant disclosure. Dispersing agents useful in the disclosure include, for example, sorbitan trioleate, oleyl alcohol, oleic acid, lecithin and corn oil. A suspension aerosol formulation can also include lubricants, preservatives, antioxidant, and/or other aerosol components.


An aerosol formulation can similarly be formulated as an emulsion. An emulsion aerosol formulation can include, for example, an alcohol such as ethanol, a surfactant, water and a propellant, as well as an agent or combination of agents of the disclosure. The surfactant used can be nonionic, anionic or cationic. One example of an emulsion aerosol formulation comprises, for example, ethanol, surfactant, water and propellant. Another example of an emulsion aerosol formulation comprises, for example, vegetable oil, glyceryl monostearate and propane.


The compounds of the disclosure can be formulated for administration as suppositories. A low melting wax, such as a mixture of triglycerides, fatty acid glycerides, Witepsol S55 (trademark of Dynamite Nobel Chemical, Germany), or cocoa butter is first melted and the active component is dispersed homogeneously, for example, by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and to solidify.


The compounds of the disclosure can be formulated for vaginal administration. Pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.


It is envisioned additionally, that the compounds of the disclosure can be attached releasably to biocompatible polymers for use in sustained release formulations on, in or attached to inserts for topical, intraocular, periocular, or systemic administration. The controlled release from a biocompatible polymer can be utilized with a water soluble polymer to form an instillable formulation, as well. The controlled release from a biocompatible polymer, such as for example, PLGA microspheres or nanospheres, can be utilized in a formulation suitable for intra ocular implantation or injection for sustained release administration, as well any suitable biodegradable and biocompatible polymer can be used.


In one aspect of the disclosure, the subject's carrier status of any of the genetic variation risk variants described herein, or genetic variants identified via other analysis methods within the genes or regulatory loci that are identified by the CNVs or SNVs described herein, can be used to help determine whether a particular treatment modality, such as any one of the above, or a combination thereof, should be administered. Whether a treatment option such as any of the above mentioned treatment options is administered can be determined based on the presence or absence of a particular genetic variation risk variant in the individual, or by monitoring expression of genes that are associated with the variants of the present disclosure. Expression levels and/or mRNA levels can thus be determined before and during treatment to monitor its effectiveness. Alternatively, or concomitantly, the status with respect to a genetic variation, and or genotype and/or haplotype status of at least one risk variant for PML presented herein can be determined before and during treatment to monitor its effectiveness. It can also be appreciated by those skilled in the art that aberrant expression levels of a gene impacted by a CNV or other mutations found as a consequence of targeted sequencing of the CNV-identified gene can be assayed or diagnostically tested for by measuring the polypeptide expression level of said aberrantly expressed gene. In another embodiment, aberrant expression levels of a gene may result from a CNV impacting a DNA sequence (e.g., transcription factor binding site) that regulates a gene whose aberrant expression level is involved in or causes PML, or other mutations found as a consequence of targeted sequencing of the CNV-identified gene regulatory sequence, can be assayed or diagnostically tested for by measuring the polypeptide expression level of the gene involved in or causative of PML. In some embodiments, a specific CNV mutation within a gene, or other specific mutations found upon targeted sequencing of a CNV-identified gene found to be involved in or causative of PML, may cause an aberrant structural change in the expressed polypeptide that results from said gene mutations and the altered polypeptide structure(s) can be assayed via various methods know to those skilled in the art.


Alternatively, biological networks or metabolic pathways related to the genes within, or associated with, the genetic variations described herein can be monitored by determining mRNA and/or polypeptide levels. This can be done for example, by monitoring expression levels of polypeptides for several genes belonging to the network and/or pathway in nucleic acid samples taken before and during treatment. Alternatively, metabolites belonging to the biological network or metabolic pathway can be determined before and during treatment. Effectiveness of the treatment is determined by comparing observed changes in expression levels/metabolite levels during treatment to corresponding data from healthy subjects.


In some embodiments, the genetic variations described herein and/or those subsequently found (e.g., via other genetic analysis methods such as sequencing) via targeted analysis of those genes initially identified by the genetic variations described herein, can be used to prevent adverse effects associated with a therapeutic agent, such as during clinical trials. For example, individuals who are carriers of at least one at-risk genetic variation can be more likely to respond negatively to a therapeutic agent, such as an immunosuppressive agent. For example, carriers of certain genetic variants may be more likely to show an adverse response to the therapeutic agent. In some embodiments, one or more of the genetic variations employed during clinical trials for a given therapeutic agent can be used in a companion diagnostic test that is administered to the patient prior to administration of the therapeutic agent to determine if the patient is likely to have a favorable or an adverse response to the therapeutic agent.


The genetic variations described herein can be used for determining whether a subject is administered a pharmaceutical agent, such as an immunosuppressive drug. Certain combinations of variants, including those described herein, but also combinations with other risk variants for PML, can be suitable for one selection of treatment options, while other variant combinations can be suitable for selection of other treatment options. Such combinations of variants can include one variant, two variants, three variants, or four or more variants, as needed to determine with clinically reliable accuracy the selection of treatment module. In another embodiment, information from testing for the genetic variations described herein, or other rare genetic variations in or near the genes described herein, may be combined with information from other types of testing (e.g., a JCV antibody test, CD62L test, or CSF IgM oligoclonal bands test) for selection of treatment options.


Kits


Kits useful in the methods of the disclosure comprise components useful in any of the methods described herein, including for example, primers for nucleic acid amplification, hybridization probes for detecting genetic variation, or other marker detection, restriction enzymes, nucleic acid probes, optionally labeled with suitable labels, allele-specific oligonucleotides, antibodies that bind to an altered polypeptide encoded by a nucleic acid of the disclosure as described herein or to a wild type polypeptide encoded by a nucleic acid of the disclosure as described herein, means for amplification of genetic variations or fragments thereof, means for analyzing the nucleic acid sequence of nucleic acids comprising genetic variations as described herein, means for analyzing the amino acid sequence of a polypeptide encoded by a genetic variation, or a nucleic acid associated with a genetic variation, etc. The kits can for example, include necessary buffers, nucleic acid primers for amplifying nucleic acids, and reagents for allele-specific detection of the fragments amplified using such primers and necessary enzymes (e.g., DNA polymerase). Additionally, kits can provide reagents for assays to be used in combination with the methods of the present disclosure, for example, reagents for use with other screening assays for PML.


In some embodiments, the disclosure pertains to a kit for assaying a nucleic acid sample from a subject to detect the presence of a genetic variation, wherein the kit comprises reagents necessary for selectively detecting at least one particular genetic variation in the genome of the individual. In some embodiments, the disclosure pertains to a kit for assaying a nucleic acid sample from a subject to detect the presence of at least one particular allele of at least one polymorphism associated with a genetic variation in the genome of the subject. In some embodiments, the reagents comprise at least one contiguous oligonucleotide that hybridizes to a fragment of the genome of the individual comprising at least genetic variation. In some embodiments, the reagents comprise at least one pair of oligonucleotides that hybridize to opposite strands of a genomic segment obtained from a subject, wherein each oligonucleotide primer pair is designed to selectively amplify a fragment of the genome of the individual that includes at least one genetic variation, or a fragment of a genetic variation. Such oligonucleotides or nucleic acids can be designed using the methods described herein. In some embodiments, the kit comprises one or more labeled nucleic acids capable of allele-specific detection of one or more specific polymorphic markers or haplotypes with a genetic variation, and reagents for detection of the label. In some embodiments, a kit for detecting SNP markers can comprise a detection oligonucleotide probe, that hybridizes to a segment of template DNA containing a SNP polymorphism to be detected, an enhancer oligonucleotide probe, detection probe, primer and/or an endonuclease, for example, as described by Kutyavin et al., (Nucleic Acid Res. 34:e128 (2006)). In other embodiments, the kit can contain reagents for detecting SNVs and/or CNVs.


In some embodiments, the DNA template is amplified by any means of the present disclosure, prior to assessment for the presence of specific genetic variations as described herein. Standard methods well known to the skilled person for performing these methods can be utilized, and are within scope of the disclosure. In one such embodiment, reagents for performing these methods can be included in the reagent kit.


In a further aspect of the present disclosure, a pharmaceutical pack (kit) is provided, the pack comprising a therapeutic agent and a set of instructions for administration of the therapeutic agent to humans screened for one or more variants of the present disclosure, as disclosed herein. The therapeutic agent can be a small molecule drug, an antibody, a peptide, an antisense or RNAi molecule, or other therapeutic molecules as described herein. In some embodiments, an individual identified as a non-carrier of at least one variant of the present disclosure is instructed to take the therapeutic agent. In one such embodiment, an individual identified as a non-carrier of at least one variant of the present disclosure is instructed to take a prescribed dose of the therapeutic agent. In some embodiments, an individual identified as a carrier of at least one variant of the present disclosure is instructed not to take the therapeutic agent. In some embodiments, an individual identified as a carrier of at least one variant of the present disclosure is instructed not to take a prescribed dose of the therapeutic agent. In some embodiments, an individual identified as a carrier of at least one variant of the present disclosure is instructed to take an agent that targets the JC Virus. For example, an individual identified as a carrier of at least one variant of the present disclosure can be instructed to take an agent that targets the JC Virus prior to or in conjunction with, taking an immunosuppressive agent.


Also provided herein are articles of manufacture, comprising a probe that hybridizes with a region of human chromosome as described herein and can be used to detect a polymorphism described herein. For example, any of the probes for detecting polymorphisms or genetic variations described herein can be combined with packaging material to generate articles of manufacture or kits. The kit can include one or more other elements including: instructions for use; and other reagents such as a label or an agent useful for attaching a label to the probe. Instructions for use can include instructions for screening applications of the probe for making a diagnosis, prognosis, or theranosis to PML in a method described herein. Other instructions can include instructions for attaching a label to the probe, instructions for performing in situ analysis with the probe, and/or instructions for obtaining a nucleic acid sample to be analyzed from a subject. In some cases, the kit can include a labeled probe that hybridizes to a region of human chromosome as described herein.


The kit can also include one or more additional reference or control probes that hybridize to the same chromosome or another chromosome or portion thereof that can have an abnormality associated with a particular endophenotype. A kit that includes additional probes can further include labels, e.g., one or more of the same or different labels for the probes. In other embodiments, the additional probe or probes provided with the kit can be a labeled probe or probes. When the kit further includes one or more additional probe or probes, the kit can further provide instructions for the use of the additional probe or probes. Kits for use in self-testing can also be provided. Such test kits can include devices and instructions that a subject can use to obtain a nucleic acid sample (e.g., buccal cells, blood) without the aid of a health care provider. For example, buccal cells can be obtained using a buccal swab or brush, or using mouthwash.


Kits as provided herein can also include a mailer (e.g., a postage paid envelope or mailing pack) that can be used to return the nucleic acid sample for analysis, e.g., to a laboratory. The kit can include one or more containers for the nucleic acid sample, or the nucleic acid sample can be in a standard blood collection vial. The kit can also include one or more of an informed consent form, a test requisition form, and instructions on how to use the kit in a method described herein. Methods for using such kits are also included herein. One or more of the forms (e.g., the test requisition form) and the container holding the nucleic acid sample can be coded, for example, with a bar code for identifying the subject who provided the nucleic acid sample.


In some embodiments, an in vitro screening test can comprise one or more devices, tools, and equipment configured to collect a nucleic acid sample from an individual. In some embodiments of an in vitro screening test, tools to collect a nucleic acid sample can include one or more of a swab, a scalpel, a syringe, a scraper, a container, and other devices and reagents designed to facilitate the collection, storage, and transport of a nucleic acid sample. In some embodiments, an in vitro screening test can include reagents or solutions for collecting, stabilizing, storing, and processing a nucleic acid sample.


Such reagents and solutions for nucleotide collecting, stabilizing, storing, and processing are well known by those of skill in the art and can be indicated by specific methods used by an in vitro screening test as described herein. In some embodiments, an in vitro screening test as disclosed herein, can comprise a microarray apparatus and reagents, a flow cell apparatus and reagents, a multiplex nucleotide sequencer and reagents, and additional hardware and software necessary to assay a nucleic acid sample for certain genetic markers and to detect and visualize certain genetic markers.


The present disclosure further relates to kits for using antibodies in the methods described herein. This includes, but is not limited to, kits for detecting the presence of a variant polypeptide in a test nucleic acid sample. One preferred embodiment comprises antibodies such as a labeled or labelable antibody and a compound or agent for detecting variant polypeptides in a nucleic acid sample, means for determining the amount or the presence and/or absence of variant polypeptide in the nucleic acid sample, and means for comparing the amount of variant polypeptide in the nucleic acid sample with a standard, as well as instructions for use of the kit. In certain embodiments, the kit further comprises a set of instructions for using the reagents comprising the kit.


Computer-Implemented Aspects


As understood by those of ordinary skill in the art, the methods and information described herein (genetic variation association with PML) can be implemented, in all or in part, as computer executable instructions on known computer readable media. For example, the methods described herein can be implemented in hardware. Alternatively, the method can be implemented in software stored in, for example, one or more memories or other computer readable medium and implemented on one or more processors. As is known, the processors can be associated with one or more controllers, calculation units and/or other units of a computer system, or implanted in firmware as desired. If implemented in software, the routines can be stored in any computer readable memory such as in RAM, ROM, flash memory, a magnetic disk, a laser disk, or other storage medium, as is also known. Likewise, this software can be delivered to a computing device via any known delivery method including, for example, over a communication channel such as a telephone line, the Internet, a wireless connection, etc., or via a transportable medium, such as a computer readable disk, flash drive, etc.


More generally, and as understood by those of ordinary skill in the art, the various steps described above can be implemented as various blocks, operations, tools, modules and techniques which, in turn, can be implemented in hardware, firmware, software, or any combination of hardware, firmware, and/or software. When implemented in hardware, some or all of the blocks, operations, techniques, etc. can be implemented in, for example, a custom integrated circuit (IC), an application specific integrated circuit (ASIC), a field programmable logic array (FPGA), a programmable logic array (PLA), etc.


Results from such genotyping can be stored in a data storage unit, such as a data carrier, including computer databases, data storage disks, or by other convenient data storage means. In certain embodiments, the computer database is an object database, a relational database or a post-relational database. Data can be retrieved from the data storage unit using any convenient data query method.


When implemented in software, the software can be stored in any known computer readable medium such as on a magnetic disk, an optical disk, or other storage medium, in a RAM or ROM or flash memory of a computer, processor, hard disk drive, optical disk drive, tape drive, etc. Likewise, the software can be delivered to a user or a computing system via any known delivery method including, for example, on a computer readable disk or other transportable computer storage mechanism.


The steps of the claimed methods can be operational with numerous other general purpose or special purpose computing system environments or configurations. Examples of well known computing systems, environments, and/or configurations that can be suitable for use with the methods or system of the claims include, but are not limited to, personal computers, server computers, hand-held or laptop devices, multiprocessor systems, microprocessor-based systems, set top boxes, programmable consumer electronics, network PCs, minicomputers, mainframe computers, distributed computing environments that include any of the above systems or devices, and the like.


The steps of the claimed method and system can be described in the general context of computer-executable instructions, such as program modules, being executed by a computer. Generally, program modules include routines, programs, objects, components, and/or data structures that perform particular tasks or implement particular abstract data types. The methods and apparatus can also be practiced in distributed computing environments where tasks are performed by remote processing devices that are linked through a communications network. In both integrated and distributed computing environments, program modules can be located in both local and remote computer storage media including memory storage devices. Numerous alternative embodiments could be implemented, using either current technology or technology developed after the filing date of this application, which would still fall within the scope of the claims defining the disclosure.


While the risk evaluation system and method, and other elements, have been described as preferably being implemented in software, they can be implemented in hardware, firmware, etc., and can be implemented by any other processor. Thus, the elements described herein can be implemented in a standard multi-purpose CPU or on specifically designed hardware or firmware such as an application-specific integrated circuit (ASIC) or other hard-wired device as desired. When implemented in software, the software routine can be stored in any computer readable memory such as on a magnetic disk, a laser disk, or other storage medium, in a RAM or ROM of a computer or processor, in any database, etc. Likewise, this software can be delivered to a user or a screening system via any known or desired delivery method including, for example, on a computer readable disk or other transportable computer storage mechanism or over a communication channel, for example, a telephone line, the internet, or wireless communication. Modifications and variations can be made in the techniques and structures described and illustrated herein without departing from the spirit and scope of the present disclosure.


Unless otherwise explained, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. The following references contain embodiments of the methods and compositions that can be used herein: The Merck Manual of Diagnosis and Therapy, 18th Edition, published by Merck Research Laboratories, 2006 (ISBN 0-911910-18-2); Benjamin Lewin, Genes IX, published by Jones & Bartlett Publishing, 2007 (ISBN-13: 9780763740634); Kendrew et al., (eds.), The Encyclopedia of Molecular Biology, published by Blackwell Science Ltd., 1994 (ISBN 0-632-02182-9); and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1-56081-569-8).


Standard procedures of the present disclosure are described, e.g., in Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (1982); Sambrook et al., Molecular Cloning: A Laboratory Manual (2 ed.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (1989); Davis et al., Basic Methods in Molecular Biology, Elsevier Science Publishing, Inc., New York, USA (1986); or Methods in Enzymology: Guide to Molecular Cloning Techniques Vol. 152, S. L. Berger and A. R. Kimmerl (eds.), Academic Press Inc., San Diego, USA (1987)). Current Protocols in Molecular Biology (CPMB) (Fred M. Ausubel, et al., ed., John Wiley and Sons, Inc.), Current Protocols in Protein Science (CPPS) (John E. Coligan, et al., ed., John Wiley and Sons, Inc.), Current Protocols in Immunology (CPI) (John E. Coligan, et al., ed. John Wiley and Sons, Inc.), Current Protocols in Cell Biology (CPCB) (Juan S. Bonifacino et al., ed., John Wiley and Sons, Inc.), Culture of Animal Cells: A Manual of Basic Technique by R. Ian Freshney, Publisher: Wiley-Liss; 5th edition (2005), and Animal Cell Culture Methods (Methods in Cell Biology, Vol. 57, Jennie P. Mather and David Barnes editors, Academic Press, 1st edition, 1998), which are all incorporated by reference herein in their entireties.


It should be understood that the following examples should not be construed as being limiting to the particular methodology, protocols, and compositions, etc., described herein and, as such, can vary. The following terms used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the embodiments disclosed herein.


Disclosed herein are molecules, materials, compositions, and components that can be used for, can be used in conjunction with, can be used in preparation for, or are products of methods and compositions disclosed herein. It is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed and while specific reference of each various individual and collective combinations and permutation of these molecules and compounds cannot be explicitly disclosed, each is specifically contemplated and described herein. For example, if a nucleotide or nucleic acid is disclosed and discussed and a number of modifications that can be made to a number of molecules including the nucleotide or nucleic acid are discussed, each and every combination and permutation of nucleotide or nucleic acid and the modifications that are possible are specifically contemplated unless specifically indicated to the contrary. This concept applies to all aspects of this application including, but not limited to, steps in methods of making and using the disclosed molecules and compositions. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the disclosed methods, and that each such combination is specifically contemplated and should be considered disclosed.


Those skilled in the art can recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the method and compositions described herein. Such equivalents are intended to be encompassed by the following claims.


It is understood that the disclosed methods and compositions are not limited to the particular methodology, protocols, and reagents described as these can vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present disclosure which can be limited only by the appended claims.


Unless defined otherwise, all technical and scientific terms used herein have the meanings that would be commonly understood by one of skill in the art in the context of the present specification.


It should be noted that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to “a nucleotide” includes a plurality of such nucleotides; reference to “the nucleotide” is a reference to one or more nucleotides and equivalents thereof known to those skilled in the art, and so forth.


The term “and/or” shall in the present context be understood to indicate that either or both of the items connected by it are involved. While preferred embodiments of the present disclosure have been shown and described herein, it can be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions can now occur to those skilled in the art without departing from the disclosure. It should be understood that various alternatives to the embodiments of the disclosure described herein can be employed in practicing the disclosure. It is intended that the following claims define the scope of the disclosure and that methods and structures within the scope of these claims and their equivalents be covered thereby.


EXAMPLES
Example 1—Experimental Approach

In the present study, a set of genes were identified, deleterious variants within which increase susceptibility to PML. The relevant genes were discovered on the basis of a combined CNV plus sequence analysis approach. Two sets of genes were compiled (see Table 6 and corresponding description):

    • A. A set based on a detailed literature review of genes involved in the immune system and JC virus biology, along with genes described in the context of PML via case reports.
    • B. A set based on the observation of rare CNVs within the PML cohort.


A non-redundant list of 419 genes was generated (see Table 6), which contains 245 curated from immune deficiency (immunodeficiency) reviews (Table 6, ‘Public_db’), 169 identified via rare CNVs using the methods described herein (Table 6, ‘PBio’), and 6 genes that were found using both methods (Table 6, ‘Both’). See Table 6 and description below for further information).


Using this set of 419 genes, it was determined whether:

    • Rare CNVs were present that might explain the susceptibility to PML;
    • Rare sequence variants (determined via whole exome sequencing analysis—WES) were present that might explain the susceptibility to PML;
    • Combinations of CNVs, SNVs and/or CNVs and SNVs might explain the susceptibility;
    • Individual variants might be present at higher frequency in the PML cohort (variant burden analysis—Tables 14, 15);
    • Total numbers of heterozygous, damaging variants were high for any specific genes (gene burden analysis—Table 13).


In all cases, due consideration was given to:

    • Pathogenic/deleterious nature of the variants observed (e.g., whether gene function was highly likely to be affected);
    • Rarity of the variants or variant combinations (e.g., those that would be expected to be present in 1% or less of the normal population were considered);
    • Ethnicity of the PML cases to account for potential frequency differences in one population subgroup vs. another. Ethnicities (e.g., ancestry) for the PML patients are reported in Table 7. For Sample_ID identifiers beginning with ‘MVGS’, ethnicities were not reported but all patients were from the USA and their ethnicities were assumed to be of European (EUR) ancestry. However, PML case MVGS811-13a is potentially of African (AFR) ancestry on the basis of common SNVs that are also found in PML cases known to be of AFR ancestry. In one embodiment, ethnic-specific frequency data from the ExAC database was used to assess relative frequencies of variants found in PML patients vs. an unselected population (ExAC subjects). ExAC ethnicities were designated as follows: African/African American (AFR), Latino (LAT, also known as AMR), East Asian (EAS), Finnish (FIN), Non-Finnish European (EUR, also known as NFE), South Asian (SAS), and Other (OTH). For some PML cases reported in Table 7, the ethnicities were alternately reported as Subsaharan, North African (MGB), Caribbean (CAR), or Hispanic (HISP). For interpretation of variants found in these patients, the assignments of ancestry using ExAC db designations were as follows: AFR=MGB or Subsaharan; LAT=CAR or HISP. Ancestry was unknown for two PML cases (PML02 and PML28) and, for frequency interpretation purposes (using ExAC db), they were assumed to be of European (EUR) ancestry.


While the primary genetic mechanism that was considered was autosomal recessive (AR) inheritance, a number of solutions were based on autosomal dominant (AD) inheritance but only in cases for which prior evidence was found that heterozygous variants in the relevant gene had previously been associated with an immune deficiency syndrome. It can be appreciated by those skilled in the art that some genes may contain both AR and AD model pathogenic variants (e.g., see Table 6 entries marked as ‘AD_AR’ in the ‘Disease_Model’ column).


For AR inheritance (˜40% of genes in Table 6 fall into this category, AR or AD_AR), the following were considered:

    • Homozygous or compound heterozygous gene-disruptive CNVs;
    • Homozygous or compound heterozygous sequence variants; i.e. single nucleotide variants (SNVs). Compound heterozygosity was only inferred when either phasing was available or one of the pairs of SNVs was itself homozygous;
    • Compound heterozygosity for a CNV and SNV. Such calls were only possible in cases for which the SNV was in trans to a deletion (e.g., DUSP16 SNV in Table 10 and the CNV in Table 1).


Example 2—Copy Number Variant (CNV) Analysis

The data presented herein was generated on the basis of a comparison of copy number variants (CNVs) identified in 2 cohorts:

    • 1) 1,005 Normal individuals (Normal Variation Engine—NVE);
    • 2) 71 Progressive Multifocal Leukoencephalopathy (PML) cases along with 6 Human Immunodeficiency Virus (HIV) cases without a diagnosis of PML (in order to aid in distinguishing germline variants vs. acquired variants that result from HIV infection). Total cohort size=77.


Genomic DNA Sample Hybridization—NVE and PML, HIV Cohorts


Genomic DNA samples from individuals within the Normal cohort (NVE ‘test’ subjects, also referred to as ‘NVE cases’ in some tables herein) and from the PML, HIV cohort (PML, HIV ‘test’ subjects) were hybridized against a single, sex-matched reference individual. Reference DNA samples were labeled with Cy5 and test subject DNA samples were labeled with Cy3. After labeling, samples were combined and co-hybridized to Agilent 1M feature oligonucleotide microarrays, design ID 021529 (Agilent Product Number G4447A) using standard conditions (array Comparative Genomic Hybridization

    • aCGH). Post-hybridization, arrays were scanned at 2 μm resolution, using Agilent's DNA microarray scanner, generating tiff images for later analysis.


All tiff images were analyzed using Agilent Feature Extraction (FE) software, with the following settings:

    • Human Genome Freeze:hg18:NCBI36:Mar2006
    • FE version: 10.7.3.1
    • Grid/design file: 021529_D_F_20091001
    • Protocol: CGH_107_Sep09


This procedure generates a variety of output files, one of which is a text-tab delimited file, containing ˜1,000,000 rows of data, each corresponding to a specific feature on the array. This *.txt file was used to perform CNV calling using DNAcopy, an open source software package implemented in R via BioConductor (http://www.bioconductor.org/packages/release/bioc/html/DNAcopy.html).


Heterozygous losses (het_loss), homozygous losses (hom_loss) or gains were determined according to a threshold log 2 ratio, which was set at:

    • hom_loss min=−1000;
    • hom_loss max=−2;
    • het_loss min=−2;
    • het_loss max=−0.5;
    • gain min=0.5;
    • gain max=1000;


With very few exceptions, all CNVs with a log 2 ratio value between −0.5 and +0.5 were not considered. All log 2 ratio values were determined according to Cy3/Cy5 (Test/Reference). A minimum probe threshold for CNV-calling was set at 2 (2 consecutive probes were sufficient to call a CNV). A CNV list was generated for each individual in the 3 cohorts (NVE, PML, and HW).


Using custom scripts, CNVs identified in the NVE and PML cohorts (many of which appeared in multiple individuals) were (separately) ‘merged’ into master lists of non-redundant CNV-subregions, according to the presence or absence of the CNV-subregion in individuals within the cohort. Using this approach, the NVE-master lists have:















7778
het_loss


653
hom_loss


4862
gain










distinct CNV-subregions, respectively. The PML+HIV cohort of 77 individuals master lists contained:















2523
het_loss


314
hom_loss


1639
gain










distinct CNV-subregions, respectively.


Those skilled in the art can appreciate that CNVs can be acquired in an individual's genome that are not inherited. Such ‘acquired CNVs’ often occur in a tissue specific manner, such as in solid tumors compared to a patient's normal tissue. In blood-derived genomic DNA samples, which are what was used for both the NVE and PML subjects in the studies described herein, acquired CNVs can be the result of blood cancers such as leukemia and lymphoma, but also due to HIV infection. Many of the PML cases in this study had HIV as their primary disease (see Table 7). In order to aid in the interpretation of acquired vs. germline CNVs, an HW sub-cohort of 6 cases was included in the primary, genome-wide CNV comparison but rare CNVs in the 6 HIV (non-PML) cases were not considered as relevant to PML susceptibility. The purpose of generating data on the 6 HIV cases was to determine whether some changes seen in PML patients who developed the disorder on a background of HIV (PML/HIV) were likely related to the underlying HIV and not the PML susceptibility itself. In other words, the HIV cases served as a general control for the large number of PML/HIV cases.


For example, consider 3 individuals within the NVE cohort with the following hypothetical CNVs:


Chr1:1-100,000; Chr1:10,001-100,000; and Chr1:1-89,999. In the master list, these would be merged into 3 distinct CNV subregions, as follows:


















CNV-subregion 1
Chr1: 1-10,000
Subjects A, C



CNV-subregion 2
Chr1: 10,001-89,999
Subjects A, B, C



CNV-subregion 3
Chr90,000: 1-100,000
Subjects A, B










Comparison of the corresponding NVE and PML master lists of CNV-subregions was performed (het_loss versus het_loss, hom_loss versus hom_loss and gain versus gain), resulting in a combined file with totals for NVE and PML for each distinct CNV-subregion in the study.


The data are subsequently curated as follows (The example calculation below was based on an original PML cohort of 80 cases, of which 6 are non-PML HIV controls and 3 PML cases that were duplicate samples. In some instances, the OR and FET values reported in Table 2 were used as ‘relative’ guidelines when considering the relevance of a CNV. In nearly all instances, a CNV was considered as a potential cause or contributing factor to PML if it was absent from the NVE database of CNVs).

    • Annotation using custom designed scripts in order to attach relevant information to each CNV region regarding overlap with known genes and exons, overlap with genes involved in the immune system and overlap with regulatory regions, including transcription factor binding sites.
    • A calculation of the odds ratio (OR) and Fishers Exact test (FET) for each CNV-subregion, according to the following formula:

      OR=(PML/(77−PML))/(NVE/(1005−NVE))
      • where:
      • PML=number of PML individuals with CNV-subregion of interest
      • NVE=number of NVE individuals with CNV-subregion of interest


As an illustrative example, consider the CNV subregion gain involving chr2:55764753-55771586, which is found in 3 individuals in the PML cohort and 1 individual in the NVE cohort (see Table 2). The OR is: (3/74)/(1/1004)=40.7


Note that, by one convention, if either of NVE or PML=0, a value of 0.5 is added to all 4 entries in the main formula above, in order to avoid dealing with infinities (see Deeks and Higgins, Statistical algorithms in Review Manager 5, Statistical Methods Group of The Cochrane Collaboration, (2010)). This has the effect of artificially lowering OR values in cases where no individuals within the NVE have the CNV. This method is applicable to all the calculations in Table 2. This method is also used when calculating the Fisher's 2-tailed Exact Test (FET) in the event that any one of the variables is zero. For convenience in analysis, the sub-cohort of 6 HIV (non-PML) cases were retained in Table 2. Therefore, the OR values reported in Table 2 are slightly different from the OR calculations for the actual number of PML cases (n=71). Using the example above for a CNV-subregion gain involving chr2:55764753-55771586, the actual OR using 71 PML cases vs. 1005 NVE subjects was: (3/68)/(1/(1004)=44.29. In some instances, a non-PML HIV control (see Table 11, identified as 3280, 3281, 3283, 3284, 3285, and 3286) is found to have a CNV of potential relevance in PML subjects. This can also impact the OR calculation. For example, for CNV-subregion loss chr19:55247874-55250186 the OR in Table 2 is listed as 17.38 but one case is a non-PML HIV control (Table 11, PML70_control=3280). For this example, the actual OR using 71 PML cases vs. 1005 NVE subjects, and excluding the non-PML HIV case, was: (4/67)/(4/(1001)=14.94.


The CNV-subregions/genes that are listed herein (e.g., in one or more of Tables 1-4), fulfill one of the following criteria:

    • Strong biology linking the gene that a CNV-subregion impacts or is near, with known immune deficiency pathways/mechanisms or biology in PML (e.g., JC virus related biology). That is, in some cases, statistical evidence is lacking but does not exclude the CNV-subregion as a candidate;
    • Statistical analysis combined with medium to strong biology (e.g., links in the peer-reviewed literature to PML, JC virus, host defense, immune deficiency, or neuropathology) without obvious biological connection (best FET in this category was 3.25E-10);


It can be appreciated by those skilled in the art that the number of PML candidate CNV-subregions, irrespective of category, may increase or decrease as additional PML cohorts are analyzed.


Example 3—Whole Exome Sequencing (WES) and Case Level Analysis

WES data was obtained on a total of 70 PML cases (non-PML HIV cases were not sequenced—they were used simply to help in the interpretation of complex CNVs observed in PML patients who also had HIV).


Variant annotation reports were further interrogated against the full set of genes detailed above. Synonymous variants and variants predicted to be modifiers (outside coding regions) were not considered. For all other variants, further filtering was performed so that only those predicted by at least one in silico prediction algorithm (e.g., Polyphen2, SIFT, MutationTaster) to be pathogenic were considered for further evaluation. Finally, only variants or variant combinations that would be expected to be present in 1% or less of the normal population were evaluated for case level analysis (Tables 7-10). Data from the Exome Aggregation Consortium (ExAC) was used to obtain ethnic-specific frequency data for variants under consideration (see, Lek et al., Nature, 17; 536(7616):285-91) (2016)).


Example 4—Description of Sequence Data

The sequence file 33655-710.101_ST25.txt contains genomic sequence information for (in the following order):

    • A. All distinct CNVs listed in Table 1;
    • B. The full genomic extent of the transcripts listed in Table 4;
    • C. Sequence variants detailed in Table 5.
    • D. The full genomic extent of the transcripts listed in Table 12


Note that:

    • 1. SEQ_ID 1-172 are the CNV sequences from Table 1;
    • 2. SEQ_ID 173-455 are the transcript sequences from Table 4;
    • 3. SEQ_ID 1000-1329 are the sequence variants from Table 5;
    • 4. SEQ_ID 1500-2177 are the transcript sequences from Table 12.


Examples of sequences submitted:









TABLE 1







Sequence entry starts:


SEQ_ID 1 = 49,653 bp CNV (het_loss) at chr1:1086119-1135772 involving


genes MIR200A, MIR200B, MIR429, TNFRSF18, TTLL10:


<210> 1


<211> 49654


<212> DNA


<213> Homo sapiens


<400> 1








cttctggggt ctaaggccag aagtgacctt tcttctcacg gaggcacccc cacatcacag
   60





gccccaagct cccaccagga gtccccaggc agcaggtttt ccaccacagc cgggaagagc
  120





cccgccttca ccacccacca ccagccaatc ccgagaccac cgaagccccc agaccgggcc
  180


. . . (sequence truncated for brevity)






gattcccgca cggccgggga cggccccagg gccttgggag cgtctgtgga cacctgtggt
49560





gtgggccgag gagctgggag ctcatctgaa cacgccagca ctcgcgcatc cacgctgctg
49620





gcggatgcct gggtttctcc actgtggggc cacg
49654


Sequence entry ends.
















TABLE 4







Sequence entry starts:


SEQ_ID 173 = MIR200B, transcript NR_029639, which is 95 bp in length:


<210> 173


<211> 95


<212> DNA


<213> Homo sapiens


<400> 173








ccagctcggg cagccgtggc catcttactg ggcagcattg gatggagtca ggtctctaat
60





actgcctggt aatgatgacg gcggagccct gcacg
95


Sequence entry ends.
















TABLE 5







Sequence entry starts:


SEQ_ID 1148 = chr9:304628   reference allele = G;


alternate allele = A


<210> 1148


<211> 40


<212> DNA


<213> Homo sapiens


<220>


<221> variant


<222> (20) . . . (20)


<223> G->A


<400> 1148








tttaaaaaga ctggatctcg aaaagatttt cacaagacgc
40


Sequence entry ends.
















TABLE 12







Sequence entry starts:


SEQ_ID 1500 = ACADM, transcript NM_000016, which is 39,313 bp in length:


<210> 1500


<211> 39313


<212> DNA


<213> Homo sapiens


<400> 1500








cgcaagtccc cccaccgttc agcgcaaccg ggccctccca gccccgccgc cgtccccctc
   60





ccccgccctg gctctctttc cgcgctgcgg tcagcctcgg cgtcccacag agagggccag
  120


. . . (sequence truncated for brevity)






gtaatagtgt atatttcttg tatttactat gatgaaaaaa ggtcgtttta attttgaatt
39240





gaataaagtt acctgttcat tttttattag atattttaaa gacttcagaa aatataaata
39300





tgaaataatt taa
39313


Sequence entry ends.









Example 5

Those skilled in the art can appreciate that genes can be impacted by acquired or germline genetic variants (e.g., CNVs), wherein each gene has the potential to contain genetic variants that are acquired (e.g., via a disease process such as HIV infection, or cancers such as leukemia and lymphoma) or present in the germ line (e.g., inherited from a parent or are de novo, i.e. not inherited from a parent). In FIG. 1, the PRKCB gene was impacted by germ line variants in 2 PML cases and acquired variants in 6 PML cases. The invention described herein is focused on detection of germline variants that are present in PML patient genomes. Therefore, no solutions/explanations for a given patient's PML was based on an acquired CNV, although another PML patient could potentially be ‘solved’ by one or two germline rare variants impacting the gene.


For this PRKCB example, no CNV-based solutions were found (an AR model was assumed), but 1 SNV solution is reported in Table 8 (het SNV, an AD model is assumed for this PML case). Further supporting evidence was assessed for the PRKCB gene by performing String analysis (high confidence=0.7, 1st shell=up to 10 interactors; string-db.org; see Szklarczyk et al., (2015), and references therein). String analysis showed that PRKCB interacts with PML-419 genes CARD11, IKBKB, and RBCK1 (see Table 6).


In FIG. 2, both TNFRSF13C and CENPM are disrupted and/or gained by a set of acquired CNV gains. Acquired CNVs can be very complex, such as the high copy number gains often identified in tumor-derived DNA samples (as compared to the patient's normal genome). In the PML gene discovery described herein, blood-derived genomic DNA obtained from several PML-diagnosed HIV patients, or PML cases with a primary disease of leukemia and lymphoma (reported as ‘Other’ in Table 7), showed complex genomic changes (e.g., gains exhibiting a dup-trip-dup pattern). In some PML cases, the acquired gains passed the log 2 ratio cutoff (>0.5) that was selected for this study, but in other PML cases the log 2 ratios for the gains were <0.5 and this data was filtered out from the main analyses that were performed to ascertain rare germline CNVs.


In one embodiment of the invention, a set of 6 non-PML HIV cases (3 African ancestry, 3 European ancestry) were used to aid in the interpretation of whether a CNV was an acquired or germline event. The non-PML ‘PML cases’ are labeled with ‘_control’ in Table 11 and correspond to ‘PML_Case_ID’ numbers 3280, 3281, 3283, 3284, 3285, and 3286. While some CNVs are reported in Tables 1 and 2 for this set of non-PML control HIV subjects, none of these genetic findings were used to nominate a gene discovered on the basis of rare CNVs (as compared to the NVE db) as a potential PML gene (PBio genes reported in Table 6). In other words, these rare CNVs were only used to aid in determining if a particular genomic region containing multiple overlapping CNVs was potentially due to an acquired genetic event. Those skilled in the art can appreciate that the set of experiments described herein do not necessarily fully rule in or out that a given genomic region contained only acquired CNVs vs. only germline CNVs (i.e. it's possible that the same region can contain an acquired CNV in one individual and a germline CNV in another).


For the CNV data shown in FIG. 2, both the TNFRSF13C and CENPM genes were included in PML-419 gene list (Table 6) on the basis of their immune or neurological related biology reported in the literature. No CNV or SNV PML solutions were found for these two genes, but String analysis (high confidence=0.7, 1st shell=up to 10 interactors) shows that TNFRSF13C interacts with PML-419 genes TRAF3 (Table 7 solution) and TNFRSF13B (Table 8 solution), as well as BTK (a known PML gene, see Table 6).



FIG. 3 shows another example of a gene that is impacted by both germline and acquired CNVs. While no PML cases were solved on the basis of the acquired or germline CNVs shown to impact the PKHD1 gene, nomination of this gene to Table 6 on the basis of its biology resulted in finding 3 potential alternate solutions (AR model) for 3 other PML cases (see Table 8). However, String analysis (high confidence=0.7, 1st shell=up to 10 interactors) did not reveal any PML-419 gene interactions with PKHD1.


Example 6

Those skilled in the art can appreciate that an AR disease model would involve ascertaining whether both alleles (for a gene or genetic locus) are impacted by a genetic variant in individuals affected by the disorder. The types of genetic variants can be SNVs, CNVs, indels, etc. In the study describe herein, if an AR disease model was invoked for a gene (see Table 6), we assessed the PML patient's CGH data for CNVs (heterozygous or homozygous) and their exome data for SNVs (heterozygous or homozygous). Thus, each patient may be solved for one of the PML-419 genes (Table 6) with one of the following scenarios: homozygous deletion, homozygous duplication (log 2 ratio will appear comparable to that typically found for triplications), homozygous SNV, compound heterozygous SNVs, compound heterozygous CNVs, or compound heterozygous SNV and CNV. Those skilled in the art know that, for an AR disease mechanism, a pathogenic SNV or CNV may have appreciable frequency in the general population (e.g., up to 1% frequency) with little to no impact on the individual's health, but when present with a second pathogenic variant on the other allele, can cause disease.



FIG. 4 shows an example of a recurrent intronic loss impacting the BMPR2 gene. Patient PML29 was found to have a homozygous deletion, whereas as patients PML58 and MVGS811-13a have a heterozygous deletion. Assuming an AR disease model, no SNV solutions were found for this gene; however, PML29 is potentially solved due to the homozygous deletion that was detected. While immune-related biology is reported for studies on BMPR2 (see Table 6), String analysis (high confidence=0.7, 1st shell=up to 10 interactors) did not reveal any PML-419 gene interactions with BMPR2.



FIG. 5 shows an example of a recurrent exonic gain that disrupts the COMMD6 gene. Two PML patients were found to have homozygous duplications of this CNV. Interestingly, while String analysis (high confidence=0.7, 1st shell=up to 10 interactors) did not reveal any PML-419 gene interactions with COMMD6, recent studies (see Table 6, PMIDs 25355947 and 27441653) show a potential link between COMMD6 and known PML gene WAS via the WASH gene.



FIG. 6 shows an example of a recurrent exonic gain that disrupts the KCTD7 gene and its right breakpoint is upstream of RABGEF1 (i.e. one or both genes may be causing/contributing to PML). A recently annotated non-coding RNA (see hg19 assembly, LOC100996437) may also be impacted by this CNV. Both genes have immune and neurological links (see Table 6) and since patient PML29 has a homozygous duplication, it was added as a PML solution in Table 7. String analysis (high confidence=0.7, 1st shell=up to 10 interactors) did not reveal any PML-419 gene interactions for either gene, but they are linked together in a joint String analysis.



FIG. 7 shows an example of a recurrent gain that disrupts FPR2 (left breakpoint) and ZNF616 (right breakpoint, gene not labeled), and other genes are fully encompassed by this CNV. There is strong supporting biology for FPR2 (see Table 6) and it is listed as a PML solution in Table 7. String analysis (high confidence=0.7, 1st shell=up to 10 interactors) did not reveal any PML-419 gene interactions for FPR2, but a joint analysis of Table 7 genes did reveal an interaction (see FIG. 13).



FIG. 8 shows an example of an exonic loss impacting the PIK3CD and PIK3CD-AS1 genes. Patient MVGS811-13a has a homozygous deletion and is reported as a solution in Table 7 based on the strong immune-related biology for PIK3CD (see Table 6). String analysis (high confidence=0.7, 1st shell=up to 10 interactors) reveals PML-419 gene interactions for PTEN and PIK3R1.


Example 7

A subset of the rare CNVs found in our PML study were located in intergenic regions. While those skilled in the art can appreciate that intergenic variants (CNVs, SNVs, etc.) can have long range effects on the expression of genes (e.g., gene regulatory elements can be located several kilobases away from the genes under their influence), in our study we assumed that intergenic CNVs were potentially impacting one or both adjacent genes if they were located <˜100 Kb away, either upstream or downstream. The ENCODE project has revealed a wealth of information, such as transcription factor binding sites, and rare CNVs that were identified in the study herein were checked for their potential impact on these sites (hg19 assembly ENCODE annotation was checked) and were often found to impact transcription factor binding sites and/or were located in conserved DNA regions.



FIG. 9 shows an intergenic, recurrent gain that is upstream of CD180. Patient MVGS995-4a has a homozygous duplication and, while not considered as a PML solution in Table 7, is potentially an alternate solution that may be causing or contributing to the patient's PML based on altered expression of CD180. The gene has immune-related biology (see Table 6) and String analysis (high confidence=0.7, 1st shell=up to 10 interactors) reveals a PML-419 gene interaction with PLCG2 (see Table 7, 2 PML cases have a solution for this gene).



FIG. 10 shows an intergenic, recurrent loss that is upstream of VDAC1. Patient PML30 has a homozygous deletion and, while not considered as a PML solution in Table 7, is potentially an alternate solution that may be causing or contributing to the patient's PML based on altered expression of VDAC1. String analysis (high confidence=0.7, 1st shell=up to 10 interactors) did not reveal any PML-419 gene interactions for VDAC1.



FIG. 11 shows an intergenic, recurrent loss that is downstream of EGR1 and ETF1. Patient PML69 has a homozygous deletion and, based on links for EGR1 to PML-419 genes (Table 6) and its proximity to EGR1 (˜4 Kb away), it was added as a potential PML solution in Table 7. String analysis (high confidence=0.7, 1st shell=up to 10 interactors) reveals PML-419 gene interactions with JUN, PTEN, and TP53), but nothing of note was found for String analysis of ETF1.



FIG. 12 shows an intergenic, recurrent loss that is upstream of ITSN2. Patient PML65 has a homozygous deletion and, based on links for ITSN2 to a known PML gene (WAS) in the PML-419 gene list (Table 6), it was added as a potential PML solution in Table 7. Interestingly, another PML case was found to have a rare homozygous SNV in ITSN2, so this gene has 2 PML solutions reported in Table 7. String analysis (high confidence=0.7, 1st shell=up to 10 interactors) did not reveal any PML-419 gene interactions.


Example 8

Pathway analyses, such as protein-protein interactions, are providing valuable insights into the underlying biology for complex diseases. While PML is a very rare disease that requires several concurrent factors (e.g., infection by the JC virus), multiple genes may be independently causing or increasing the risk of developing this neurodegenerative disorder based on the presence of a genetic variant in a given gene (e.g., a heterozygous variant wherein one deleterious variant is present on the maternally or paternally inherited allele, a homozygous variant wherein the same deleterious variant is present on both alleles, or compound heterozygous variants wherein a pair of deleterious variants are present but one is found on the maternally inherited allele and the other is found on the paternally inherited allele). As hypothesized, presence of an immune deficiency genetic disorder was another prerequisite. Indeed, in the PML study described herein, 43 genes were proposed as solutions for 61 of 71 PML cases (see Table 7) that were assessed using array CGH and whole exome sequencing. Numerous algorithms and associated databases have been developed to investigate molecular pathways, such as String (see, Szklarczyk et al., (2015), and references therein).



FIG. 13 shows an example of String analysis performed on the 43 genes considered as PML solutions on the basis of an AD or AR disease model. A series of interactions were found for 21 of 43 genes, and in several instances this included interactions for genes implicated in 2 or more PML cases that are reported in Table 7 (9 cases for TNFRSF11A, 4 cases for PLCG2, 3 cases for ZAP70 and NOD2, and 2 PML cases for TICAM1).


Example 9

To determine the likelihood that a randomly selected individual would harbor one of the variants described herein, the following analysis was performed: For each variant or combination of variants, the ethnic-specific frequency quoted in Table 7 was used to determine the probability that a randomly selected individual of the same ethnicity would be expected not to harbor the variant or combination of variants. The product of all such probabilities was calculated (e.g., the probability that a randomly selected individual would not harbor any of the variants) and subtracted from 1, yielding the probability that a random individual would harbor at least one of the variants. It was found that, for HIV cases, the probability of a random individual harboring at least one of the variants was ˜5%, which is consistent with the pre-HAART risk of PML in the context of HIV. For non-HIV cases (mostly MS/NTZ), the risk was ˜1%, which, again, is consistent with the risk of PML in MS/NTZ, especially after long-term therapy.


These analyses support the notion that the frequencies of the variants identified as relevant to PML risk are consistent with the actual observed risks for unselected individuals. The analyses are predicated on the reasonable assumption that there is no PML-relevant connection with the risk of developing HIV (an acquired infection) and/or MS (e.g., this implies that treatment of healthy individuals with Natalizumab, for example, would result in similar risks of PML). Any deviations (e.g., variants found in a slightly higher number of normal individuals than expected according to the numbers actually observed to be affected by PML) may be due to: penetrance (e.g., not everyone with the variants will be at maximal risk of PML); the assumption that individuals with MS, HIV and other underlying conditions represented a normal (e.g., with respect to PML risk) cross-section of the general population, prior to developing the underlying disorders HIV, MS etc; and under ascertainment of PML, even in patients with HIV, MS/NTZ.


Example 10—Tables Referenced in this Study








TABLE 1







CNVs of interest in this study















Original CNV
Original
Original

PML

SEQ


Chr
Start
CNV Stop
CNV Size
CNV Type
Case ID
RefSeq Gene Symbol
ID

















1
1086119
1135772
49653
het loss
3009
MIR200A, MIR200B, MIR429, TNFRSF18, TTLL10
1


1
9634094
9635206
1112
hom loss
3009
PIK3CD
2


1
12018512
12032581
14069
gain
3205

3


1
19593401
19602807
9406
het_loss
3203
CAPZB
4


1
21695957
21700243
4286
het loss
3161

5


1
24364786
24391166
26380
gain
3199
IFNLR1
6


1
28666669
28737671
71002
gain
3161
PHACTR4, RCC1, SNHG3
7


1
49372054
49380088
8034
het_loss
3145
AGBL4
8


1
153816159
153827698
11539
het loss
3168

9


1
205607255
205610341
3086
gain
3007

10


1
215760485
215762451
1966
het_loss
3117
GPATCH2
11


1
215866737
215869900
3163
het_loss
3151
GPATCH2
12


2
10352668
10356083
3415
het_loss
3007

13


2
24457024
24462631
5607
hom loss
3204

14


2
38468717
38471950
3233
het loss
3175

15


2
38516138
38524237
8099
het_loss
3151

16


2
38726517
38731845
5328
het_loss
3159

17


2
40620890
40624089
3199
het_loss
3202

18


2
46631006
46643501
12495
gain
3145
RHOQ
19


2
55764753
55790559
25806
gain
3143
PNPT1
20


2
55764753
55790559
25806
gain
3193
PNPT1
20


2
55764753
55790559
25806
gain
3282
PNPT1
20


2
55764753
55790559
25806
gain
3143
PNPT1
20


2
55764753
55790559
25806
gain
3193
PNPT1
20


2
55764753
55790559
25806
gain
3282
PNPT1
20


2
55764753
55790559
25806
gain
3143
PNPT1
20


2
55764753
55790559
25806
gain
3193
PNPT1
20


2
55764753
55790559
25806
gain
3282
PNPT1
20


2
71190677
71200120
9443
het_loss
3175
MCEE
21


2
71190677
71200120
9443
het loss
3175
MCEE
21


2
71191311
71200120
8809
het loss
3204
MCEE
22


2
71198108
71200120
2012
het loss
3143
MCEE
23


2
71190677
71200120
9443
het_loss
3175
MCEE
21


2
71198108
71200120
2012
het_loss
3193
MCEE
23


2
71198108
71200120
2012
het loss
3200
MCEE
23


2
71191311
71200120
8809
het loss
3204
MCEE
22


2
74773432
74913493
140061
gain
3118
HK2
24


2
105418748
105435274
16526
het loss
3193
FHL2
25


2
110182348
110210249
27901
gain
3174
MALL, MIR4267, MIR4436B1, MIR4436B2
26


2
127823042
127828410
5368
het_loss
3273

27


2
134911636
134914254
2618
het loss
3273
MGAT5
28


2
203005216
203019933
14717
het loss
3009
BMPR2
29


2
203005216
203019933
14717
het_loss
3192
BMPR2
29


2
203005216
203019933
14717
hom_loss
3152
BMPR2
29


2
230212897
230216339
3442
het loss
3154
DNER
30


3
122979920
122994402
14482
gain
3202
IQCB1
31


4
26565071
26566345
1274
het loss
3010
STIM2
32


4
26565071
26566345
1274
het_loss
3125
STIM2
32


4
26565071
26566345
1274
het_loss
3168
STIM2
32


4
26565071
26566345
1274
het loss
3282
STIM2
32


4
26565071
26566345
1274
het loss
3284
STIM2
32


4
26565071
26566345
1274
hom loss
3273
STIM2
32


4
54838623
54873909
35286
gain
3153
PDGFRA
33


4
90791460
90843887
52427
gain
3168

35


4
90800863
90808258
7395
het loss
3009

35


4
90800863
90808258
7395
het loss
3284

35


5
45331278
46150784
819506
gain
3157
HCN1
36


5
49771219
49774457
3238
gain
3273
EMB
37


5
66619415
66636116
16701
gain
3010

38


5
78480194
78497296
17102
gain
3205

39


5
78497296
78531091
33795
gain
3132

40


5
78497296
78521408
24112
gain
3185

41


5
78497296
78531091
33795
gain
3132

40


5
78497296
78521408
24112
gain
3185

41


5
78500552
78526637
26085
gain
3205

42


5
78497296
78531091
33795
gain
3132

40


5
78500552
78526637
26085
gain
3205

42


5
78497296
78531091
33795
gain
3132

40


5
83490494
83495169
4675
het_loss
3204
EDIL3
43


5
133372071
133379727
7656
hom_loss
3153

44


5
137836466
137843309
6843
hom loss
3279

45


5
150159466
150202601
43135
het loss
3117

46


5
150159466
150204134
44668
het_loss
3180

47


5
150159466
150202601
43135
het_loss
3199

46


5
150159466
150204134
44668
het loss
3278

47


5
150159466
150202601
43135
het_loss
3117

46


5
150159466
150204134
44668
het_loss
3180

47


5
150159466
150202601
43135
het loss
3199

46


5
150159466
150204134
44668
het loss
3278

47


5
150159466
150202601
43135
het_loss
3117

46


5
150159466
150204134
44668
het_loss
3180

47


5
150159466
150202601
43135
het loss
3199

46


5
150159466
150204134
44668
het loss
3278

47


5
150185190
150201145
15955
hom_loss
3009

48


5
150185190
150201145
15955
hom_loss
3143

48


5
150185190
150202601
17411
hom loss
3152

49


5
150185190
150202601
17411
hom loss
3154

49


5
150185190
150202601
17411
hom_loss
3193

49


5
150159466
150201145
41679
hom_loss
3196

50


5
150185190
150201145
15955
hom loss
3281

48


5
150185190
150201145
15955
hom loss
3009

48


5
150185190
150201145
15955
hom_loss
3143

48


5
150185190
150202601
17411
hom_loss
3152

49


5
150185190
150202601
17411
hom loss
3154

49


5
150185190
150202601
17411
hom loss
3193

49


5
150159466
150201145
41679
hom_loss
3196

50


5
150185190
150201145
15955
hom_loss
3281

48


5
150185190
150202601
17411
hom loss
3152

49


5
150185190
150202601
17411
hom loss
3154

49


5
150185190
150202601
17411
hom loss
3193

49


5
150185190
150204134
18944
het loss
3132

51


5
150159466
150204134
44668
het_loss
3180

47


5
150202601
150204134
1533
het_loss
3196

52


5
150191322
150204134
12812
het loss
3273

53


5
150185190
150204134
18944
het loss
3277

51


5
150159466
150204134
44668
het_loss
3278

47


5
150185190
150204134
18944
het_loss
3280

51


5
150185190
150204134
18944
het loss
3282

51


5
179590681
179626660
35979
het loss
3172
MAPK9
54


6
2882577
2947403
64826
het loss
3196
DKFZP686I15217, NQO2, SERPINB6
55


6
2964646
2966011
1365
het_loss
3193
HTATSF1P2, NQO2
56


6
51766024
51773250
7226
het_loss
3167
PKHD1
57


6
51952217
51969378
17161
gain
3127
PKHD1
58


6
51952217
51969378
17161
gain
3127
PKHD1
58


6
51953476
51965723
12247
gain
3205
PKHD1
59


6
51952217
51969378
17161
gain
3127
PKHD1
58


6
74396294
74404837
8543
het_loss
3009
SLC17A5
60


6
74396294
74398409
2115
het loss
3160
SLC17A5
61


6
74396294
74404837
8543
het loss
3009
SLC17A5
60


6
86416979
86431527
14548
het loss
3197

62


6
91131823
91135670
3847
het_loss
3171

63


6
107882367
107890605
8238
het loss
3201
PDSS2
64


6
166418511
166422386
3875
het loss
3125

65


6
166418511
166422386
3875
het_loss
3163

65


6
166418511
166422386
3875
het_loss
3192

65


6
166418511
166422386
3875
het loss
3193

65


6
166418511
166422386
3875
het loss
3194

65


6
166418511
166422386
3875
het_loss
3200

65


6
166418511
166422386
3875
het_loss
3205

65


6
166418511
166422386
3875
het loss
3280

65


6
166418511
166422386
3875
het loss
3281

65


6
166418511
166422386
3875
het_loss
3284

65


6
166418511
166422386
3875
hom_loss
3009

65


6
166418511
166422386
3875
hom loss
3152

65


6
166418511
166422386
3875
hom loss
3175

65


7
65741238
65768682
27444
gain
3152
KCTD7
66


7
65741238
65768682
27444
gain
3202
KCTD7
66


7
157174966
157177843
2877
het loss
3009
PTPRN2
67


7
157425841
157496238
70397
gain
3189
PTPRN2
68


7
158000082
158024569
24487
het_loss
3279
PTPRN2
69


7
158000082
158024569
24487
het_loss
3279
PTPRN2
69


7
158000082
158024569
24487
het loss
3279
MIR595, PTPRN2
69


8
23103186
23125443
22257
het_loss
3140
TNFRSF10A
70


8
39914488
39919594
5106
het_loss
3126
IDO2
71


8
79905654
79910286
4632
het loss
3159

72


8
99790200
99799839
9639
het loss
3006
STK3
73


8
102049360
102064431
15071
het loss
3173

74


8
102049360
102064431
15071
het_loss
3175

74


8
102049360
102064431
15071
het_loss
3282

74


9
571398
584647
13249
het_loss
3006
KANK1
75


9
571398
584647
13249
het_loss
3006
KANK1
75


9
580722
598488
17766
het loss
3200
KANK1
76


9
580722
598488
17766
het loss
3282
KANK1
76


9
580722
598488
17766
het_loss
3200
KANK1
76


9
580722
598488
17766
het_loss
3282
KANK1
76


9
634039
637589
3550
het_loss
3273
KANK1
77


9
634039
637589
3550
het loss
3282
KANK1
77


9
74050088
74059447
9359
het loss
3165
GDA
78


9
93140394
93447826
307432
gain
3198
AUH, MIR3163, MIR3910-1, MIR3910-2,
79








NFIL3



9
118564159
118575633
11474
gain
3193
ASTN2
80


9
118612694
118664593
51899
het_loss
3144
ASTN2
81


9
119220847
119233078
12231
gain
3005

82


10
899657
1071401
171744
gain
3161
GTPBP4, IDI2, IDI2-AS1, LARP4B
83


10
76217585
76411591
194006
gain
3179
KAT6B
84


10
116000069
116004388
4319
gain
3010
VWA2
85


11
14677012
14689025
12013
het_loss
3199
PDE3B
86


11
34608313
34615878
7565
het_loss
3117
EHF
87


11
62382087
62398462
16375
het loss
3205
SLC3A2
88


11
76631014
76643625
12611
het loss
3193
GDPD4
89


12
11616557
12422129
805572
het_loss
3126
ETV6
90


12
12435301
12778142
342841
het loss
3126
APOLD1, CDKN1B, CREBL2, DUSP16, GPR19,
91








LOH12CR1



12
12968705
12971310
2605
gain
3127

92


12
91786998
94313682
2526684
het_loss
3126
EEA1, LOC643339
93


12
91786998
94313682
2526684
het loss
3126
LOC643339, MRPL42, NUDT4, NUDT4P1, SOCS2,
93








SOCS2-AS1, UBE2N



12
91786998
94313682
2526684
het_loss
3126
CCDC41, CRADD, PLXNC1
93


12
111061085
111064486
3401
het loss
3004
TRAFD1
94


13
40939924
41026908
86984
gain
3140
RGCC
95


13
75006025
75016304
10279
gain
3009
COMMD6
96


13
75006025
75016304
10279
gain
3152
COMMD6
96


13
91811087
91814369
3282
het_loss
3143
GPC5
97


13
91811087
91811118
31
hom loss
3173
GPC5
98


13
110754499
110778301
23802
gain
3006
ARHGEF7, TEX29
99


14
20021118
20055469
34351
gain
3205
RNASE10
100


14
20426824
20481852
55028
hom loss
3200
ECRP, RNASE3
101


14
20430810
20490129
59319
het_loss
3192
ECRP
102


14
20430810
20490129
59319
het_loss
3192

102


14
20430810
20490129
59319
het_loss
3192

102


14
21096689
21105611
8922
het loss
3125

103


14
21096689
21105611
8922
het loss
3175

103


14
21096689
21105611
8922
het_loss
3194

103


14
21096689
21105611
8922
het_loss
3204

103


14
21096689
21105611
8922
het_loss
3273

103


14
21120750
21125513
4763
gain
3143

104


14
21120750
21125513
4763
gain
3173

104


14
60901636
60909492
7856
het_loss
3193
PRKCH
105


14
60912874
60921269
8395
het_loss
3174
PRKCH
106


14
63937192
63944459
7267
gain
3205
MTHFD1
107


14
95754535
95759056
4521
het loss
3009
BDKRB2
108


14
95754535
95759056
4521
het loss
3173
BDKRB2
108


14
95754535
95759056
4521
het_loss
3202
BDKRB2
108


15
66065925
66082418
16493
het_loss
3010

109


15
70432627
70443017
10390
gain
3169
HEXA
110


15
75096101
75128723
32622
gain
3200
PSTPIP1
111


15
75101524
75115806
14282
gain
3132
PSTPIP1
112


15
75096101
75128723
32622
gain
3200
PSTPIP1
111


15
75105789
75115806
10017
gain
3127
PSTPIP1
113


15
75101524
75115806
14282
gain
3132
PSTPIP1
112


15
75105789
75115806
10017
gain
3199
PSTPIP1
113


15
75096101
75128723
32622
gain
3200
PSTPIP1
111


15
75105789
75115806
10017
gain
3279
PSTPIP1
113


15
75105789
75115806
10017
gain
3127
PSTPIP1
113


15
75101524
75115806
14282
gain
3132
PSTPIP1
112


15
75105789
75115806
10017
gain
3199
PSTPIP1
113


15
75096101
75128723
32622
gain
3200
PSTPIP1
111


15
75105789
75115806
10017
gain
3279
PSTPIP1
113


15
75096101
75128723
32622
gain
3200
PSTPIP1
111


15
88999998
89016848
16850
het loss
3172

114


16
6823677
6932753
109076
het loss
3126
RBFOX1
115


16
6823677
6932753
109076
het_loss
3126
RBFOX1
115


16
6942078
6945539
3461
gain
3173
RBFOX1
116


16
6942078
6945539
3461
gain
3175
RBFOX1
116


16
6942078
6945539
3461
gain
3282
RBFOX1
116


16
23842653
23848772
6119
het loss
3198
PRKCB
117


16
23892842
23903495
10653
gain
3199
PRKCB
118


16
23892842
23903495
10653
gain
3199
PRKCB
118


16
23893969
23908248
14279
gain
3205
PRKCB
119


16
23893969
23908248
14279
gain
3205
PRKCB
119


16
69044235
69050151
5916
gain
3174
FUK
120


16
69044235
69050151
5916
gain
3185
FUK
120


16
69052450
69081640
29190
het loss
3197
COG4, FUK
121


16
70653499
70665447
11948
gain
3143
HPR
122


16
70653499
70665447
11948
gain
3152
HPR
122


16
70653499
70665447
11948
gain
3192
HPR
122


16
70653499
70665447
11948
gain
3200
HPR
122


16
70653499
70665447
11948
gain
3282
HPR
122


16
70653499
70665447
11948
gain
3284
HPR
122


17
69341925
70202523
860598
gain
3183
BTBD17, C17orf77, CD300A, CD300C, CD300E,
123








CD300LB, CD300LD, CD300LF, DNAI2,









GPR142, GPRC5C, KIF19, MGC16275, RAB37,









RPL38, TTYH2



17
75608151
75615433
7282
het loss
3144
TBC1D16
124


17
75608151
75615433
7282
het loss
3152
TBC1D16
124


17
75608151
75615433
7282
het_loss
3163
TBC1D16
124


17
75608151
75611602
3451
het_loss
3192
TBC1D16
125


17
75608151
75615433
7282
het loss
3200
TBC1D16
124


17
75608151
75611602
3451
het loss
3204
TBC1D16
125


17
75608151
75611602
3451
het loss
3284
TBC1D16
125


17
75608151
75611602
3451
hom_loss
3009
TBC1D16
125


17
75611602
75615433
3831
hom_loss
3175
TBC1D16
126


17
75608151
75615433
7282
het loss
3144
TBC1D16
124


17
75608151
75615433
7282
het loss
3152
TBC1D16
124


17
75608151
75615433
7282
het_loss
3163
TBC1D16
124


17
75608151
75615433
7282
het_loss
3200
TBC1D16
124


17
76241510
76267844
26334
gain
3205
RPTOR
127


17
76247305
76265683
18378
gain
3127
RPTOR
128


17
76241510
76267844
26334
gain
3205
RPTOR
127


17
76241510
76267844
26334
gain
3205
RPTOR
127


18
9985530
10125331
139801
gain
3175

129


18
12764095
12781985
17890
gain
3191
PTPN2
130


18
27026203
27029351
3148
het loss
3125

131


18
27026203
27029351
3148
het loss
3143

131


18
27026203
27029351
3148
het_loss
3175

131


18
42537949
42663605
125656
gain
3125
PIAS2, ST8SIA5
132


18
46917195
46945018
27823
het loss
3161

133


18
59457622
59465699
8077
het loss
3145
SERPINB4
134


19
3270755
3291144
20389
gain
3205

135


19
46386511
46388364
1853
hom_loss
3175

136


19
52496536
52501292
4756
gain
3124

137


19
55247874
55252420
4546
het loss
3163
FLJ26850
138


19
55247874
55252420
4546
het loss
3173
FLJ26850
138


19
55247874
55252420
4546
het loss
3192
FLJ26850
138


19
55247874
55252420
4546
het_loss
3200
FLJ26850
138


19
55247874
55252420
4546
het_loss
3280
FLJ26850
138


19
55247874
55252420
4546
het loss
3163
FLJ26850
138


19
55247874
55252420
4546
het loss
3173
FLJ26850
138


19
55247874
55252420
4546
het_loss
3192
FLJ26850
138


19
55250187
55252420
2233
het_loss
3194
FLJ26850
139


19
55247874
55252420
4546
het_loss
3200
FLJ26850
138


19
55247874
55252420
4546
het loss
3280
FLJ26850
138


19
55250187
55252420
2233
hom loss
3175
FLJ26850
139


19
55250187
55252420
2233
hom_loss
3202
FLJ26850
139


19
56964168
57308449
344281
gain
3155
FPR2, FPR3, ZNF350, ZNF432, ZNF577, ZNF613,
140








ZNF614, ZNF615, ZNF649, ZNF841



19
56964168
57308449
344281
gain
3157
FPR2, FPR3, ZNF350, ZNF432, ZNF577, ZNF613,
140








ZNF614, ZNF615, ZNF649, ZNF841



19
59013780
59023850
10070
het loss
3117
NLRP12
141


19
59249279
59251831
2552
hom loss
3160
VSTM1
142


19
59249279
59251831
2552
hom_loss
3164
VSTM1
142


19
59250742
59251831
1089
hom_loss
3117
VSTM1
143


19
59249279
59251831
2552
hom loss
3160
VSTM1
142


19
59249279
59251831
2552
hom loss
3164
VSTM1
142


19
59250742
59251831
1089
hom_loss
3277
VSTM1
143


20
17844577
17954650
110073
gain
3166
MGME1, OVOL2, SNORD17, SNX5
144


20
42706680
42711434
4754
het_loss
3125
ADA
145


21
15234620
15312960
78340
gain
3009
NRIP1
146


21
29643302
29647950
4648
het loss
3202
BACH1
147


21
44634707
44666832
32125
gain
3200
TRPM2
148


21
44634707
44641658
6951
gain
3205
TRPM2
149


21
44634707
44671482
36775
gain
3279
TRPM2
150


21
44637544
44669596
32052
gain
3127
TRPM2
151


21
44637544
44657372
19828
gain
3185
TRPM2
152


21
44634707
44666832
32125
gain
3200
TRPM2
148


21
44634707
44641658
6951
gain
3205
TRPM2
149


21
44634707
44671482
36775
gain
3279
TRPM2
150


21
44637544
44669596
32052
gain
3127
TRPM2
151


21
44637544
44657372
19828
gain
3185
TRPM2
152


21
44634707
44666832
32125
gain
3200
TRPM2
148


21
44634707
44671482
36775
gain
3279
TRPM2
150


21
44643974
44657372
13398
het loss
3161
TRPM2
153


21
44637544
44669596
32052
gain
3127
TRPM2
151


21
44637544
44657372
19828
gain
3185
TRPM2
152


21
44634707
44666832
32125
gain
3200
TRPM2
148


21
44643974
44657372
13398
gain
3205
TRPM2
153


21
44634707
44671482
36775
gain
3279
TRPM2
150


21
44637544
44669596
32052
gain
3127
TRPM2
151


21
44634707
44666832
32125
gain
3200
TRPM2
148


21
44634707
44671482
36775
gain
3279
TRPM2
150


21
44637544
44669596
32052
gain
3127
TRPM2
151


21
44634707
44666832
32125
gain
3200
TRPM2
148


21
44660199
44681194
20995
gain
3205
TRPM2
154


21
44634707
44671482
36775
gain
3279
TRPM2
150


21
44637544
44669596
32052
gain
3127
TRPM2
151


21
44660199
44681194
20995
gain
3205
TRPM2
154


21
44634707
44671482
36775
gain
3279
TRPM2
150


21
44660199
44681194
20995
gain
3205
TRPM2
154


21
44634707
44671482
36775
gain
3279
TRPM2
150


21
44660199
44681194
20995
gain
3205
TRPM2
154


21
45348895
45354820
5925
het_loss
3179
ADARB1
155


22
37689058
37715385
26327
gain
3169
APOBEC3A, APOBEC3A B, APOBEC3B
156


22
39257585
39261621
4036
het loss
3005
MKL1
157


22
40642402
40655210
12808
gain
3205
TNFRSF13C
158


22
40655820
40673250
17430
gain
3185

159


22
40655820
40675788
19968
gain
3205

160


22
40659633
40671866
12233
gain
3127

161


22
40655820
40673250
17430
gain
3185

159


22
40655820
40675788
19968
gain
3205

160


22
40659633
40671866
12233
gain
3127
CENPM
161


22
40655820
40673250
17430
gain
3185
CENPM
159


22
40663050
40668079
5029
gain
3190
CENPM
162


22
40663050
40668079
5029
gain
3202
CENPM
162


22
40655820
40675788
19968
gain
3205
CENPM
160


22
40659633
40671866
12233
gain
3127
CENPM
161


22
40655820
40673250
17430
gain
3185
CENPM
159


22
40655820
40675788
19968
gain
3205
CENPM
160


22
40655820
40673250
17430
gain
3185
CENPM
159


22
40655820
40675788
19968
gain
3205
CENPM
160


22
40655820
40675788
19968
gain
3205

160


23
232907
244684
11777
het loss
3007
PPP2R3B
163


23
7585301
7830994
245693
gain
3172

164


23
7585301
7830994
245693
gain
3172
VCX
164


23
7769323
7779354
10031
het_loss
3132

165


23
6465033
8093113
1628080
het_loss
3171

166


23
7769323
7779354
10031
het loss
3204

165


23
7585301
7830994
245693
gain
3172

164


23
7585301
7830994
245693
gain
3172

164


23
6465033
8093113
1628080
het_loss
3171
MIR651, PNPLA4
166


23
7585301
7830994
245693
gain
3172
PNPLA4
164


23
48358646
48408854
50208
het loss
3009

167


23
64710574
64725828
15254
gain
3125

168


23
73083877
73086192
2315
hom_loss
3193
JPX
169


23
73083877
73086192
2315
hom_loss
3200
JPX
169


23
122337025
122340879
3854
hom_loss
3125
GRIA3
170


23
148452844
148461889
9045
het loss
3163

171


23
148452844
148461889
9045
het loss
3205

171


23
148452844
148461889
9045
hom_loss
3144

171


23
148452844
148461889
9045
hom_loss
3193

171


23
149901706
149904265
2559
gain
3117
HMGB3
172


23
149901706
149904265
2559
gain
3118
HMGB3
172









Table 1 lists all CNVs of interest, obtained as described in the text, with the exception that, for each entry, the original CNV start and stop positions are noted, along with original CNV size, type (heterozygous loss, homozygous loss or gain), Case_ID and gene annotation (for the CNV-subregion NOT original CNV). The final column contains SEQ_ID numbers. Standard chromosomal numbering used by those skilled in the art is used in Table 1 for the autosomal chromosomes (1-22) but, for convenience with analysis methods, chromosome X is designated as chromosome 23 herein. All coordinates are based on hg18.









TABLE 2







CNV -subregions of interest in this study































CNV



CNV
CNV
CNV








Sub-



Sub-
Sub-
Sub-

PML

Exon




region



region
region
region
CNV
Case

over-
NVE
PML


No


Chr
Start
Stop
Size
Type
ID
RefSeq Gene Symbol
lap
cases
cases
FET
OR
(SRN)






















1
1086119
1135772
49653
het_loss
3009
MIR200A, MIR200B,
Y
0
1
0.005115965
39.43
1








MIR429, TNFRSF18,














TTLL10








1
9634094
9635206
1112
hom_loss
3009
PIK3CD
Y
0
1
0.005115965
39.43
2


1
12018512
12032581
14069
gain
3205

N
0
1
0.005115965
39.43
3


1
19593401
19602807
9406
het_loss
3203
CAPZB
N
0
1
0.005115965
39.43
4


1
21698753
21700243
1490
het_loss
3161

N
0
1
0.005115965
39.43
5


1
24364786
24391166
26380
gain
3199
IFNLR1
Y
0
1
0.005115965
39.43
6


1
28666669
28737671
71002
gain
3161
PHACTR4, RCC1,
Y
0
1
0.005115965
39.43
7








SNHG3








1
49372054
49380088
8034
het loss
3145
AGBL4
N
0
1
0.005115965
39.43
8


1
153816159
153827698
11539
het loss
3168

N
0
1
0.005115965
39.43
9


1
205607255
205610341
3086
gain
3007

N
0
1
0.005115965
39.43
10


1
215760485
215762451
1966
het_loss
3117
GPATCH2
N
0
1
0.005115965
39.43
11


1
215866737
215869900
3163
het_loss
3151
GPATCH2
N
0
1
0.005115965
39.43
12


2
10352668
10356083
3415
het loss
3007

N
0
1
0.005115965
39.43
13


2
24457024
24462631
5607
hom_loss
3204

N
0
1
0.005115965
39.43
14


2
38468717
38471950
3233
het loss
3175

N
0
1
0.005115965
39.43
15


2
38516138
38524237
8099
het loss
3151

N
0
1
0.005115965
39.43
16


2
38726517
38731845
5328
het_loss
3159

N
0
1
0.005115965
39.43
17


2
40620890
40624089
3199
het_loss
3202

N
0
1
0.005115965
39.43
18


2
46631006
46643501
12495
gain
3145
RHOQ
N
0
1
0.005115965
39.43
19


2
55764753
55771586
6833
gain
3143
PNPT1
Y
1
3
0.001318303
40.7
20


2
55764753
55771586
6833
gain
3193
PNPT1
Y
1
3
0.001318303
40.7
21


2
55764753
55771586
6833
gain
3282
PNPT1
Y
1
3
0.001318303
40.7
22


2
55771587
55772965
1378
gain
3143
PNPT1
N
2
3
0.003126725
20.33
23


2
55771587
55772965
1378
gain
3193
PNPT1
N
2
3
0.003126725
20.33
24


2
55771587
55772965
1378
gain
3282
PNPT1
N
2
3
0.003126725
20.33
25


2
55772966
55790559
17593
gain
3143
PNPT1
Y
1
3
0.001318303
40.7
26


2
55772966
55790559
17593
gain
3193
PNPT1
Y
1
3
0.001318303
40.7
27


2
55772966
55790559
17593
gain
3282
PNPT1
Y
1
3
0.001318303
40.7
28


2
71190677
71191310
633
het loss
3175
MCEE
Y
0
1
0.005115965
39.43
29


2
71191311
71198107
6796
het loss
3175
MCEE
N
1
2
0.014314826
26.77
30


2
71191311
71198107
6796
het loss
3204
MCEE
N
1
2
0.014314826
26.77
31


2
71198108
71200120
2012
het_loss
3143
MCEE
N
2
5
3.02E−05
34.83
32


2
71198108
71200120
2012
het_loss
3175
MCEE
N
2
5
3.02E−05
34.83
33


2
71198108
71200120
2012
het loss
3193
MCEE
N
2
5
3.02E−05
34.83
34


2
71198108
71200120
2012
het loss
3200
MCEE
N
2
5
3.02E−05
34.83
35


2
71198108
71200120
2012
het_loss
3204
MCEE
N
2
5
3.02E−05
34.83
36


2
74827730
74913493
85763
gain
3118
HK2
Y
0
1
0.005115965
39.43
37


2
105418748
105435274
16526
het loss
3193
FHL2
Y
0
1
0.005115965
39.43
38


2
110182348
110210249
27901
gain
3174
MALL, MIR4267,
Y
2
1
0.198831257
6.6
39








MIR4436B1, MIR4436B2








2
127823042
127828410
5368
het_loss
3273

N
0
1
0.005115965
39.43
40


2
134911636
134914254
2618
het_loss
3273
MGAT5
N
0
1
0.005115965
39.43
41


2
203005216
203019933
14717
het loss
3009
BMPR2
N
2
2
0.02731135 
13.37
42


2
203005216
203019933
14717
het loss
3192
BMPR2
N
2
2
0.02731135 
13.37
43


2
203005216
203019933
14717
hom_loss
3152
BMPR2
N
0
1
0.005115965
39.43
44


2
230212897
230216339
3442
het loss
3154
DNER
N
0
1
0.005115965
39.43
45


3
122979920
122994402
14482
gain
3202
IQCB1
Y
0
1
0.005115965
39.43
46


4
26565071
26566345
1274
het_loss
3010
STIM2
N
85
5
0.671895631
0.75
47


4
26565071
26566345
1274
het_loss
3125
STIM2
N
85
5
0.671895631
0.75
48


4
26565071
26566345
1274
het loss
3168
STIM2
N
85
5
0.671895631
0.75
49


4
26565071
26566345
1274
het loss
3282
STIM2
N
85
5
0.671895631
0.75
50


4
26565071
26566345
1274
het loss
3284
STIM2
N
85
5
0.671895631
0.75
51


4
26565071
26566345
1274
hom_loss
3273
STIM2
N
1
1
0.13732578 
13.21
52


4
54838623
54873909
35286
gain
3153
PDGFRA
Y
0
1
0.005115965
39.43
53


4
90791460
90843887
52427
gain
3168

N
0
1
0.005115965
39.43
54


4
90800863
90808258
7395
het_loss
3009

N
0
2
0.005115965
66.59
55


4
90800863
90808258
7395
het loss
3284

N
0
2
0.005115965
66.59
56


5
45331278
45785151
453873
gain
3157
HCN1
Y
0
1
0.005115965
39.43
57


5
49771219
49774457
3238
gain
3273
EMB
Y
0
1
0.005115965
39.43
58


5
66619415
66636116
16701
gain
3010

N
0
1
0.005115965
39.43
59


5
78480194
78497296
17102
gain
3205

N
0
1
0.005115965
39.43
60


5
78497296
78500551
3255
gain
3132

N
0
2
0.005115965
66.59
61


5
78497296
78500551
3255
gain
3185

N
0
2
0.005115965
66.59
62


5
78500552
78521408
20856
gain
3132

N
0
3
2.49E−05
94.48
63


5
78500552
78521408
20856
gain
3185

N
0
3
2.49E−05
94.48
64


5
78500552
78521408
20856
gain
3205

N
0
3
2.49E−05
94.48
65


5
78521409
78526637
5228
gain
3132

N
0
2
0.005115965
66.59
66


5
78521409
78526637
5228
gain
3205

N
0
2
0.005115965
66.59
67


5
78526638
78531091
4453
gain
3132

N
0
1
0.005115965
39.43
68


5
83490494
83495169
4675
het_loss
3204
EDIL3
N
0
1
0.005115965
39.43
69


5
133372071
133379727
7656
hom_loss
3153

N
0
1
0.005115965
39.43
70


5
137836466
137843309
6843
hom_loss
3279

N
1
1
0.13732578 
13.21
71


5
150159466
150161037
1571
het loss
3117

N
15
4
0.040487703
3.62
72


5
150159466
150161037
1571
het_loss
3180

N
15
4
0.040487703
3.62
73


5
150159466
150161037
1571
het_loss
3199

N
15
4
0.040487703
3.62
74


5
150159466
150161037
1571
het loss
3278

N
15
4
0.040487703
3.62
75


5
150161038
150181399
20361
het loss
3117

N
14
4
0.033744017
3.88
76


5
150161038
150181399
20361
het_loss
3180

N
14
4
0.033744017
3.88
77


5
150161038
150181399
20361
het_loss
3199

N
14
4
0.033744017
3.88
78


5
150161038
150181399
20361
het_loss
3278

N
14
4
0.033744017
3.88
79


5
150181400
150185189
3789
het loss
3117

N
13
4
0.027710312
4.18
80


5
150181400
150185189
3789
het loss
3180

N
13
4
0.027710312
4.18
81


5
150181400
150185189
3789
het_loss
3199

N
13
4
0.027710312
4.18
82


5
150181400
150185189
3789
het loss
3278

N
13
4
0.027710312
4.18
83


5
150185190
150191626
6436
hom_loss
3009

N
6
7
8.59E−06
16.65
84


5
150185190
150191626
6436
hom_loss
3143

N
6
7
8.59E−06
16.65
85


5
150185190
150191626
6436
hom_loss
3152

N
6
7
8.59E−06
16.65
86


5
150185190
150191626
6436
hom_loss
3154

N
6
7
8.59E−06
16.65
87


5
150185190
150191626
6436
hom_loss
3193

N
6
7
8.59E−06
16.65
88


5
150185190
150191626
6436
hom_loss
3196

N
6
7
8.59E−06
16.65
89


5
150185190
150191626
6436
hom_loss
3281

N
6
7
8.59E−06
16.65
90


5
150191627
150201145
9518
hom_loss
3009

N
6
7
8.59E−06
16.65
91


5
150191627
150201145
9518
hom_loss
3143

N
6
7
8.59E−06
16.65
92


5
150191627
150201145
9518
hom_loss
3152

N
6
7
8.59E−06
16.65
93


5
150191627
150201145
9518
hom_loss
3154

N
6
7
8.59E−06
16.65
94


5
150191627
150201145
9518
hom_loss
3193

N
6
7
8.59E−06
16.65
95


5
150191627
150201145
9518
hom_loss
3196

N
6
7
8.59E−06
16.65
96


5
150191627
150201145
9518
hom_loss
3281

N
6
7
8.59E−06
16.65
97


5
150201146
150202601
1455
hom_loss
3152

N
1
3
0.001318303
40.7
98


5
150201146
150202601
1455
hom_loss
3154

N
1
3
0.001318303
40.7
99


5
150201146
150202601
1455
hom_loss
3193

N
1
3
0.001318303
40.7
100


5
150202602
150204134
1532
het loss
3132

N
51
8
0.062987683
2.17
101


5
150202602
150204134
1532
het loss
3180

N
51
8
0.062987683
2.17
102


5
150202602
150204134
1532
het loss
3196

N
51
8
0.062987683
2.17
103


5
150202602
150204134
1532
het_loss
3273

N
51
8
0.062987683
2.17
104


5
150202602
150204134
1532
het_loss
3277

N
51
8
0.062987683
2.17
105


5
150202602
150204134
1532
het loss
3278

N
51
8
0.062987683
2.17
106


5
150202602
150204134
1532
het loss
3280

N
51
8
0.062987683
2.17
107


5
150202602
150204134
1532
het loss
3282

N
51
8
0.062987683
2.17
108


5
179590681
179626660
35979
het_loss
3172
MAPK9
Y
0
1
0.005115965
39.43
109


6
2882577
2947403
64826
het_loss
3196
DKFZP686I15217,
Y
0
1
0.005115965
39.43
110








NQO2, SERPINB6








6
2964646
2966011
1365
het_loss
3193
HTATSF1P2, NQO2
Y
0
1
0.005115965
39.43
111


6
51766024
51773250
7226
het_loss
3167
PKHD1
N
0
1
0.005115965
39.43
112


6
51952217
51953475
1258
gain
3127
PKHD1
N
0
1
0.005115965
39.43
113


6
51953476
51965723
12247
gain
3127
PKHD1
N
0
2
0.005115965
66.59
114


6
51953476
51965723
12247
gain
3205
PKHD1
N
0
2
0.005115965
66.59
115


6
51965724
51969378
3654
gain
3127
PKHD1
N
0
1
0.005115965
39.43
116


6
74396294
74398409
2115
het_loss
3009
SLC17A5
N
0
2
0.005115965
66.59
117


6
74396294
74398409
2115
het_loss
3160
SLC17A5
N
0
2
0.005115965
66.59
118


6
74398410
74404837
6427
het loss
3009
SLC17A5
Y
0
1
0.005115965
39.43
119


6
86416979
86431527
14548
het loss
3197

N
0
1
0.005115965
39.43
120


6
91131823
91135670
3847
het_loss
3171

N
0
1
0.005115965
39.43
121


6
107882367
107890605
8238
het_loss
3201
PDSS2
Y
0
1
0.005115965
39.43
122


6
166418511
166422386
3875
het loss
3125

N
11
10
3.49E−07
13.49
123


6
166418511
166422386
3875
het loss
3163

N
11
10
3.49E−07
13.49
124


6
166418511
166422386
3875
het_loss
3192

N
11
10
3.49E−07
13.49
125


6
166418511
166422386
3875
het_loss
3193

N
11
10
3.49E−07
13.49
126


6
166418511
166422386
3875
het loss
3194

N
11
10
3.49E−07
13.49
127


6
166418511
166422386
3875
het loss
3200

N
11
10
3.49E−07
13.49
128


6
166418511
166422386
3875
het loss
3205

N
11
10
3.49E−07
13.49
129


6
166418511
166422386
3875
het_loss
3280

N
11
10
3.49E−07
13.49
130


6
166418511
166422386
3875
het_loss
3281

N
11
10
3.49E−07
13.49
131


6
166418511
166422386
3875
het loss
3284

N
11
10
3.49E−07
13.49
132


6
166418511
166422386
3875
hom_loss
3009

N
0
3
2.49E−05
94.48
133


6
166418511
166422386
3875
hom_loss
3152

N
0
3
2.49E−05
94.48
134


6
166418511
166422386
3875
hom_loss
3175

N
0
3
2.49E−05
94.48
135


7
65741238
65768682
27444
gain
3152
KCTD7
Y
0
2
0.005115965
66.59
136


7
65741238
65768682
27444
gain
3202
KCTD7
Y
0
2
0.005115965
66.59
137


7
157174966
157177843
2877
het_loss
3009
PTPRN2
N
0
1
0.005115965
39.43
138


7
157425841
157496238
70397
gain
3189
PTPRN2
N
1
1
0.13732578 
13.21
139


7
158000082
158007892
7810
het loss
3279
PTPRN2
N
1
1
0.13732578 
13.21
140


7
158007893
158010672
2779
het_loss
3279
PTPRN2
N
5
1
0.358539546
2.63
141


7
158010673
158024569
13896
het_loss
3279
MIR595, PTPRN2
Y
1
1
0.13732578 
13.21
142


8
23103186
23125443
22257
het_loss
3140
TNFRSF10A
Y
0
1
0.005115965
39.43
143


8
39914488
39919594
5106
het loss
3126
IDO2
N
0
1
0.005115965
39.43
144


8
79905654
79910286
4632
het loss
3159

N
0
1
0.005115965
39.43
145


8
99790200
99799839
9639
het_loss
3006
STK3
N
0
1
0.005115965
39.43
146


8
102049360
102064431
15071
het_loss
3173

N
0
3
2.49E−05
94.48
147


8
102049360
102064431
15071
het_loss
3175

N
0
3
2.49E−05
94.48
148


8
102049360
102064431
15071
het loss
3282

N
0
3
2.49E−05
94.48
149


9
571398
580721
9323
het loss
3006
KANK1
N
2
1
0.198831257
6.6
150


9
580722
584647
3925
het_loss
3006
KANK1
N
3
3
0.005933668
13.54
151


9
580722
584647
3925
het_loss
3200
KANK1
N
3
3
0.005933668
13.54
152


9
580722
584647
3925
het loss
3282
KANK1
N
3
3
0.005933668
13.54
153


9
584648
598488
13840
het loss
3200
KANK1
N
2
2
0.02731135 
13.37
154


9
584648
598488
13840
het loss
3282
KANK1
N
2
2
0.02731135 
13.37
155


9
634039
637589
3550
het_loss
3273
KANK1
N
0
2
0.005115965
66.59
156


9
634039
637589
3550
het_loss
3282
KANK1
N
0
2
0.005115965
66.59
157


9
74050088
74059447
9359
het loss
3165
GDA
Y
0
1
0.005115965
39.43
158


9
93140394
93447826
307432
gain
3198
AUH, MIR3163, MIR3910-
Y
0
1
0.005115965
39.43
159








1, MIR3910-














2, NFIL3








9
118564159
118575633
11474
gain
3193
ASTN2
N
0
1
0.005115965
39.43
160


9
118657526
118664593
7067
het_loss
3144
ASTN2
N
0
1
0.005115965
39.43
161


9
119220847
119233078
12231
gain
3005

N
0
1
0.005115965
39.43
162


10
899657
1071401
171744
gain
3161
GTPBP4, IDI2, IDI2-
Y
0
1
0.005115965
39.43
163








AS1, LARP4B








10
76217585
76411591
194006
gain
3179
KAT6B
Y
0
1
0.005115965
39.43
164


10
116000069
116004388
4319
gain
3010
VWA2
Y
0
1
0.005115965
39.43
165


11
14677012
14689025
12013
het_loss
3199
PDE3B
N
0
1
0.005115965
39.43
166


11
34608313
34615878
7565
het_loss
3117
EHF
Y
0
1
0.005115965
39.43
167


11
62382087
62398462
16375
het loss
3205
SLC3A2
Y
0
1
0.005115965
39.43
168


11
76631014
76643625
12611
het loss
3193
GDPD4
Y
0
1
0.005115965
39.43
169


12
11616557
12114030
497473
het_loss
3126
ETV6
Y
0
1
0.005115965
39.43
170


12
12438904
12778142
339238
het_loss
3126
APOLD1, CDKN1B,
Y
0
1
0.005115965
39.43
171








CREBL2, DUSP16,














GPR19, LOH12CR1








12
12968705
12971310
2605
gain
3127

N
0
1
0.005115965
39.43
172


12
91845527
92201342
355815
het_loss
3126
EEA1, LOC643339
Y
0
1
0.005115965
39.43
173


12
92215898
92567120
351222
het_loss
3126
LOC643339, MRPL42,
Y
0
1
0.005115965
39.43
174








NUDT4, NUDT4P1,














SOCS2, SOCS2-














AS1, UBE2N








12
92568362
93307172
738810
het loss
3126
CCDC41, CRADD,
Y
0
1
0.005115965
39.43
175








PLXNC1








12
111061085
111064486
3401
het_loss
3004
TRAFD1
Y
0
1
0.005115965
39.43
176


13
40939924
41026908
86984
gain
3140
RGCC
Y
0
1
0.005115965
39.43
177


13
75006025
75016304
10279
gain
3009
COMMD6
Y
0
2
0.005115965
66.59
178


13
75006025
75016304
10279
gain
3152
COMMD6
Y
0
2
0.005115965
66.59
179


13
91811087
91814369
3282
het loss
3143
GPC5
N
1
1
0.13732578 
13.21
180


13
91811087
91811118
31
hom_loss
3173
GPC5
N
0
1
0.005115965
39.43
181


13
110754499
110778301
23802
gain
3006
ARHGEF7, TEX29
Y
0
1
0.005115965
39.43
182


14
20021118
20055469
34351
gain
3205
RNASE10
Y
0
1
0.005115965
39.43
183


14
20426824
20481852
55028
hom loss
3200
ECRP, RNASE3
Y
0
1
0.005115965
39.43
184


14
20430810
20458350
27540
het loss
3192
ECRP
Y
3
1
0.256004559
4.39
185


14
20458351
20481852
23501
het_loss
3192

N
4
1
0.309147091
3.29
186


14
20481853
20490129
8276
het_loss
3192

N
1
1
0.13732578 
13.21
187


14
21096689
21105611
8922
het_loss
3125

N
0
5
1.16E−07
152.56
188


14
21096689
21105611
8922
het_loss
3175

N
0
5
1.16E−07
152.56
189


14
21096689
21105611
8922
het loss
3194

N
0
5
1.16E−07
152.56
190


14
21096689
21105611
8922
het loss
3204

N
0
5
1.16E−07
152.56
191


14
21096689
21105611
8922
het_loss
3273

N
0
5
1.16E−07
152.56
192


14
21120750
21125513
4763
gain
3143

N
1
2
0.014314826
26.77
193


14
21120750
21125513
4763
gain
3173

N
1
2
0.014314826
26.77
194


14
60901636
60909492
7856
het loss
3193
PRKCH
N
0
1
0.005115965
39.43
195


14
60912874
60921269
8395
het loss
3174
PRKCH
N
0
1
0.005115965
39.43
196


14
63937192
63944459
7267
gain
3205
MTHFD1
Y
0
1
0.005115965
39.43
197


14
95754535
95759056
4521
het_loss
3009
BDKRB2
N
0
3
2.49E−05
94.48
198


14
95754535
95759056
4521
het loss
3173
BDKRB2
N
0
3
2.49E−05
94.48
199


14
95754535
95759056
4521
het loss
3202
BDKRB2
N
0
3
2.49E−05
94.48
200


15
66065925
66082418
16493
het loss
3010

N
0
1
0.005115965
39.43
201


15
70432627
70443017
10390
gain
3169
HEXA
Y
0
1
0.005115965
39.43
202


15
75096101
75101523
5422
gain
3200
PSTPIP1
Y
0
1
0.005115965
39.43
203


15
75101524
75105788
4264
gain
3132
PSTPIP1
Y
0
2
0.005115965
66.59
204


15
75101524
75105788
4264
gain
3200
PSTPIP1
Y
0
2
0.005115965
66.59
205


15
75105789
75109086
3297
gain
3127
PSTPIP1
Y
0
5
1.16E−07
152.56
206


15
75105789
75109086
3297
gain
3132
PSTPIP1
Y
0
5
1.16E−07
152.56
207


15
75105789
75109086
3297
gain
3199
PSTPIP1
Y
0
5
1.16E−07
152.56
208


15
75105789
75109086
3297
gain
3200
PSTPIP1
Y
0
5
1.16E−07
152.56
209


15
75105789
75109086
3297
gain
3279
PSTPIP1
Y
0
5
1.16E−07
152.56
210


15
75109087
75115806
6719
gain
3127
PSTPIP1
Y
1
5
9.14E−06
69.72
211


15
75109087
75115806
6719
gain
3132
PSTPIP1
Y
1
5
9.14E−06
69.72
212


15
75109087
75115806
6719
gain
3199
PSTPIP1
Y
1
5
9.14E−06
69.72
213


15
75109087
75115806
6719
gain
3200
PSTPIP1
Y
1
5
9.14E−06
69.72
214


15
75109087
75115806
6719
gain
3279
PSTPIP1
Y
1
5
9.14E−06
69.72
215


15
75115807
75117798
1991
gain
3200
PSTPIP1
Y
1
1
0.13732578 
13.21
216


15
88999998
89016848
16850
het_loss
3172

N
0
1
0.005115965
39.43
217


16
6823677
6884976
61299
het_loss
3126
RBFOX1
N
0
1
0.005115965
39.43
218


16
6886815
6896330
9515
het loss
3126
RBFOX1
N
0
1
0.005115965
39.43
219


16
6942078
6945539
3461
gain
3173
RBFOX1
N
1
3
0.001318303
40.7
220


16
6942078
6945539
3461
gain
3175
RBFOX1
N
1
3
0.001318303
40.7
221


16
6942078
6945539
3461
gain
3282
RBFOX1
N
1
3
0.001318303
40.7
222


16
23844022
23848772
4750
het_loss
3198
PRKCB
N
7
1
0.447101793
1.88
223


16
23892842
23893968
1126
gain
3199
PRKCB
N
0
1
0.005115965
39.43
224


16
23893969
23903495
9526
gain
3199
PRKCB
N
0
2
0.005115965
66.59
225


16
23893969
23903495
9526
gain
3205
PRKCB
N
0
2
0.005115965
66.59
226


16
23903496
23908248
4752
gain
3205
PRKCB
Y
0
1
0.005115965
39.43
227


16
69047888
69050151
2263
gain
3174
FUK
N
0
2
0.005115965
66.59
228


16
69047888
69050151
2263
gain
3185
FUK
N
0
2
0.005115965
66.59
229


16
69052450
69081640
29190
het loss
3197
COG4, FUK
Y
0
1
0.005115965
39.43
230


16
70653499
70665447
11948
gain
3143
HPR
Y
0
6
1.16E−07
182.82
231


16
70653499
70665447
11948
gain
3152
HPR
Y
0
6
1.16E−07
182.82
232


16
70653499
70665447
11948
gain
3192
HPR
Y
0
6
1.16E−07
182.82
233


16
70653499
70665447
11948
gain
3200
HPR
Y
0
6
1.16E−07
182.82
234


16
70653499
70665447
11948
gain
3282
HPR
Y
0
6
1.16E−07
182.82
235


16
70653499
70665447
11948
gain
3284
HPR
Y
0
6
1.16E−07
182.82
236


17
69341925
70202523
860598
gain
3183
BTBD17, C17orf77,
Y
1
1
0.13732578 
13.21
237








CD300A, CD300C,














CD300E, CD300LB,














CD300LD, CD300LF,














DNAI2, GPR142,














GPRC5C, KIF19,














MGC16275, RAB37,














RPL38, TTYH2








17
75608151
75611602
3451
het_loss
3144
TBC1D16
N
1
7
5.37E−08
100.4
238


17
75608151
75611602
3451
het loss
3152
TBC1D16
N
1
7
5.37E−08
100.4
239


17
75608151
75611602
3451
het loss
3163
TBC1D16
N
1
7
5.37E−08
100.4
240


17
75608151
75611602
3451
het loss
3192
TBC1D16
N
1
7
5.37E−08
100.4
241


17
75608151
75611602
3451
het_loss
3200
TBC1D16
N
1
7
5.37E−08
100.4
242


17
75608151
75611602
3451
het_loss
3204
TBC1D16
N
1
7
5.37E−08
100.4
243


17
75608151
75611602
3451
het loss
3284
TBC1D16
N
1
7
5.37E−08
100.4
244


17
75608151
75611602
3451
hom_loss
3009
TBC1D16
N
0
1
0.005115965
39.43
245


17
75611602
75615433
3831
hom_loss
3175
TBC1D16
N
0
1
0.005115965
39.43
246


17
75611603
75615433
3830
het_loss
3144
TBC1D16
N
1
4
0.000112689
55.01
247


17
75611603
75615433
3830
het_loss
3152
TBC1D16
N
1
4
0.000112689
55.01
248


17
75611603
75615433
3830
het loss
3163
TBC1D16
N
1
4
0.000112689
55.01
249


17
75611603
75615433
3830
het loss
3200
TBC1D16
N
1
4
0.000112689
55.01
250


17
76241510
76247304
5794
gain
3205
RPTOR
N
0
1
0.005115965
39.43
251


17
76247305
76265683
18378
gain
3127
RPTOR
N
0
2
0.005115965
66.59
252


17
76247305
76265683
18378
gain
3205
RPTOR
N
0
2
0.005115965
66.59
253


17
76265684
76267844
2160
gain
3205
RPTOR
N
0
1
0.005115965
39.43
254


18
9985530
10125331
139801
gain
3175

N
0
1
0.005115965
39.43
255


18
12764095
12781985
17890
gain
3191
PTPN2
Y
0
1
0.005115965
39.43
256


18
27026203
27029351
3148
het loss
3125

N
0
3
2.49E−05
94.48
257


18
27026203
27029351
3148
het loss
3143

N
0
3
2.49E−05
94.48
258


18
27026203
27029351
3148
het_loss
3175

N
0
3
2.49E−05
94.48
259


18
42537949
42663605
125656
gain
3125
PIAS2, ST8SIA5
Y
0
1
0.005115965
39.43
260


18
46917195
46945018
27823
het loss
3161

N
0
1
0.005115965
39.43
261


18
59457622
59465699
8077
het loss
3145
SERPINB4
Y
0
1
0.005115965
39.43
262


19
3270755
3291144
20389
gain
3205

N
0
1
0.005115965
39.43
263


19
46386511
46388364
1853
hom_loss
3175

N
0
1
0.005115965
39.43
264


19
52496536
52501292
4756
gain
3124

N
0
1
0.005115965
39.43
265


19
55247874
55250186
2312
het_loss
3163
FLJ26850
N
4
5
0.000161709
17.38
266


19
55247874
55250186
2312
het_loss
3173
FLJ26850
N
4
5
0.000161709
17.38
267


19
55247874
55250186
2312
het loss
3192
FLJ26850
N
4
5
0.000161709
17.38
268


19
55247874
55250186
2312
het loss
3200
FLJ26850
N
4
5
0.000161709
17.38
269


19
55247874
55250186
2312
het loss
3280
FLJ26850
N
4
5
0.000161709
17.38
270


19
55250187
55252420
2233
het_loss
3163
FLJ26850
N
4
6
1.80E−05
21.15
271


19
55250187
55252420
2233
het_loss
3173
FLJ26850
N
4
6
1.80E−05
21.15
272


19
55250187
55252420
2233
het loss
3192
FLJ26850
N
4
6
1.80E−05
21.15
273


19
55250187
55252420
2233
het loss
3194
FLJ26850
N
4
6
1.80E−05
21.15
274


19
55250187
55252420
2233
het loss
3200
FLJ26850
N
4
6
1.80E−05
21.15
275


19
55250187
55252420
2233
het_loss
3280
FLJ26850
N
4
6
1.80E−05
21.15
276


19
55250187
55252420
2233
hom_loss
3175
FLJ26850
N
0
2
0.005115965
66.59
277


19
55250187
55252420
2233
hom_loss
3202
FLJ26850
N
0
2
0.005115965
66.59
278


19
56964168
57308449
344281
gain
3155
FPR2, FPR3, ZNF350,
Y
3
2
0.043434433
8.91
279








ZNF432, ZNF577,














ZNF613, ZNF614,














ZNF615, ZNF649,














ZNF841








19
56964168
57308449
344281
gain
3157
FPR2, FPR3, ZNF350,
Y
3
2
0.043434433
8.91
280








ZNF432, ZNF577,














ZNF613, ZNF614,














ZNF615, ZNF649,














ZNF841








19
59016855
59023850
6995
het_loss
3117
NLRP12
Y
0
1
0.005115965
39.43
281


19
59249279
59250741
1462
hom_loss
3160
VSTM1
N
37
2
1      
0.7
282


19
59249279
59250741
1462
hom_loss
3164
VSTM1
N
37
2
1      
0.7
283


19
59250742
59251831
1089
hom_loss
3117
VSTM1
N
38
4
0.533838399
1.39
284


19
59250742
59251831
1089
hom_loss
3160
VSTM1
N
38
4
0.533838399
1.39
285


19
59250742
59251831
1089
hom_loss
3164
VSTM1
N
38
4
0.533838399
1.39
286


19
59250742
59251831
1089
hom_loss
3277
VSTM1
N
38
4
0.533838399
1.39
287


20
17844577
17954650
110073
gain
3166
MGME1, OVOL2,
Y
0
1
0.005115965
39.43
288








SNORD17, SNX5








20
42706680
42711434
4754
het loss
3125
ADA
N
0
1
0.005115965
39.43
289


21
15237071
15312960
75889
gain
3009
NRIP1
Y
0
1
0.005115965
39.43
290


21
29643302
29647950
4648
het loss
3202
BACH1
Y
0
1
0.005115965
39.43
291


21
44634707
44637543
2836
gain
3200
TRPM2
Y
1
3
0.001318303
40.7
292


21
44634707
44637543
2836
gain
3205
TRPM2
Y
1
3
0.001318303
40.7
293


21
44634707
44637543
2836
gain
3279
TRPM2
Y
1
3
0.001318303
40.7
294


21
44637544
44641658
4114
gain
3127
TRPM2
Y
1
5
9.14E−06
69.72
295


21
44637544
44641658
4114
gain
3185
TRPM2
Y
1
5
9.14E−06
69.72
296


21
44637544
44641658
4114
gain
3200
TRPM2
Y
1
5
9.14E−06
69.72
297


21
44637544
44641658
4114
gain
3205
TRPM2
Y
1
5
9.14E−06
69.72
298


21
44637544
44641658
4114
gain
3279
TRPM2
Y
1
5
9.14E−06
69.72
299


21
44641659
44643973
2314
gain
3127
TRPM2
Y
1
4
0.000112689
55.01
300


21
44641659
44643973
2314
gain
3185
TRPM2
Y
1
4
0.000112689
55.01
301


21
44641659
44643973
2314
gain
3200
TRPM2
Y
1
4
0.000112689
55.01
302


21
44641659
44643973
2314
gain
3279
TRPM2
Y
1
4
0.000112689
55.01
303


21
44643974
44657372
13398
het_loss
3161
TRPM2
Y
1
1
0.13732578 
13.21
304


21
44643975
44657372
13397
gain
3127
TRPM2
Y
0
5
1.16E−07
152.56
305


21
44643975
44657372
13397
gain
3185
TRPM2
Y
0
5
1.16E−07
152.56
306


21
44643975
44657372
13397
gain
3200
TRPM2
Y
0
5
1.16E−07
152.56
307


21
44643975
44657372
13397
gain
3205
TRPM2
Y
0
5
1.16E−07
152.56
308


21
44643975
44657372
13397
gain
3279
TRPM2
Y
0
5
1.16E−07
152.56
309


21
44657373
44660198
2825
gain
3127
TRPM2
Y
0
3
2.49E−05
94.48
310


21
44657373
44660198
2825
gain
3200
TRPM2
Y
0
3
2.49E−05
94.48
311


21
44657373
44660198
2825
gain
3279
TRPM2
Y
0
3
2.49E−05
94.48
312


21
44660199
44666832
6633
gain
3127
TRPM2
Y
0
4
2.49E−05
123.12
313


21
44660199
44666832
6633
gain
3200
TRPM2
Y
0
4
2.49E−05
123.12
314


21
44660199
44666832
6633
gain
3205
TRPM2
Y
0
4
2.49E−05
123.12
315


21
44660199
44666832
6633
gain
3279
TRPM2
Y
0
4
2.49E−05
123.12
316


21
44666833
44669596
2763
gain
3127
TRPM2
Y
0
3
2.49E−05
94.48
317


21
44666833
44669596
2763
gain
3205
TRPM2
Y
0
3
2.49E−05
94.48
318


21
44666833
44669596
2763
gain
3279
TRPM2
Y
0
3
2.49E−05
94.48
319


21
44669597
44671482
1885
gain
3205
TRPM2
Y
0
2
0.005115965
66.59
320


21
44669597
44671482
1885
gain
3279
TRPM2
Y
0
2
0.005115965
66.59
321


21
44671483
44681194
9711
gain
3205
TRPM2
Y
0
1
0.005115965
39.43
322


21
45348895
45354820
5925
het_loss
3179
ADARB1
N
0
1
0.005115965
39.43
323


22
37689058
37715385
26327
gain
3169
APOBEC3A,
Y
0
1
0.005115965
39.43
324








APOBEC3A_B,














APOBEC3B








22
39257585
39261621
4036
het loss
3005
MKL1
N
0
1
0.005115965
39.43
325


22
40642402
40655210
12808
gain
3205
TNFRSF13C
Y
0
1
0.005115965
39.43
326


22
40655820
40659632
3812
gain
3185

N
0
2
0.005115965
66.59
327


22
40655820
40659632
3812
gain
3205

N
0
2
0.005115965
66.59
328


22
40659633
40663049
3416
gain
3127

N
0
3
2.49E−05
94.48
329


22
40659633
40663049
3416
gain
3185

N
0
3
2.49E−05
94.48
330


22
40659633
40663049
3416
gain
3205

N
0
3
2.49E−05
94.48
331


22
40663050
40668079
5029
gain
3127
CENPM
Y
0
5
1.16E−07
152.56
332


22
40663050
40668079
5029
gain
3185
CENPM
Y
0
5
1.16E−07
152.56
333


22
40663050
40668079
5029
gain
3190
CENPM
Y
0
5
1.16E−07
152.56
334


22
40663050
40668079
5029
gain
3202
CENPM
Y
0
5
1.16E−07
152.56
335


22
40663050
40668079
5029
gain
3205
CENPM
Y
0
5
1.16E−07
152.56
336


22
40668080
40671866
3786
gain
3127
CENPM
Y
0
3
2.49E−05
94.48
337


22
40668080
40671866
3786
gain
3185
CENPM
Y
0
3
2.49E−05
94.48
338


22
40668080
40671866
3786
gain
3205
CENPM
Y
0
3
2.49E−05
94.48
339


22
40671867
40673250
1383
gain
3185
CENPM
Y
0
2
0.005115965
66.59
340


22
40671867
40673250
1383
gain
3205
CENPM
Y
0
2
0.005115965
66.59
341


22
40673251
40675788
2537
gain
3205

N
0
1
0.005115965
39.43
342


23
232907
234429
1522
het loss
3007
PPP2R3B
N
0
1
0.005115965
39.43
343


23
7585301
7769322
184021
gain
3172

N
5
1
0.358539546
2.63
344


23
7769323
7773949
4626
gain
3172
VCX
Y
7
1
0.447101793
1.88
345


23
7773982
7779354
5372
het_loss
3132

N
0
3
2.49E−05
94.48
346


23
7773982
7779354
5372
het loss
3171

N
0
3
2.49E−05
94.48
347


23
7773982
7779354
5372
het loss
3204

N
0
3
2.49E−05
94.48
348


23
7773982
7779353
5371
gain
3172

N
5
1
0.358539546
2.63
349


23
7779354
7815400
36046
gain
3172

N
6
1
0.404443314
2.19
350


23
7779355
8093113
313758
het_loss
3171
MIR651, PNPLA4
Y
0
1
0.005115965
39.43
351


23
7815401
7830994
15593
gain
3172
PNPLA4
Y
7
1
0.447101793
1.88
352


23
48358646
48408854
50208
het loss
3009

N
0
1
0.005115965
39.43
353


23
64710574
64725828
15254
gain
3125

N
0
1
0.005115965
39.43
354


23
73083877
73086192
2315
hom_loss
3193
JPX
N
1
2
0.014314826
26.77
355


23
73083877
73086192
2315
hom_loss
3200
JPX
N
1
2
0.014314826
26.77
356


23
122337025
122340879
3854
hom_loss
3125
GRIA3
N
0
1
0.005115965
39.43
357


23
148452844
148461889
9045
het_loss
3163

N
7
2
0.129983268
3.8
358


23
148452844
148461889
9045
het loss
3205

N
7
2
0.129983268
3.8
359


23
148459108
148461889
2781
hom_loss
3144

N
0
2
0.005115965
66.59
360


23
148459108
148461889
2781
hom_loss
3193

N
0
2
0.005115965
66.59
361


23
149901706
149902701
995
gain
3117
HMGB3
Y
0
2
0.005115965
66.59
362


23
149901706
149902701
995
gain
3118
HMGB3
Y
0
2
0.005115965
66.59
363









Table 2 is identical to Table 1, with a number of exceptions. Firstly, the CNV coordinates listed refer to the actual CNV-subregions found to be unique or significantly different between the disease and normal cohorts, as opposed to Table 1, which lists the original CNVs. Secondly, an extra column details whether genic CNV-subregions of interest overlap an exon or not. Third and fourth, 2 extra columns detail the number of normal cases and the number of disease cases that harbor the relevant CNV-subregion. Finally, 2 columns report Fisher's 2-tailed Exact Test (FET) and the odds ratio (OR). Standard chromosomal numbering used by those skilled in the art is used in Table 2 for the autosomal chromosomes (1-22) but, for convenience with analysis methods, chromosome X is designated as chromosome 23 herein. All coordinates are in hg18.









TABLE 3







A non-redundant list of genes listed in Table 2

















Gene


RefSeq Gene
Exon
NCBI
Gene

#


Symbol
overlap
Gene ID
Description
RefSeq_Summary
(GN)















ADARB1
intronic
104
double-stranded
This gene encodes the enzyme responsible for pre-mRNA editing of the
2





RNA-specific
glutamate receptor subunit B by site-specific deamination of adenosines. Studies






editase 1
in rat found that this enzyme acted on its own pre-mRNA molecules to convert






isoform 1
an AA dinucleotide to an AI dinucleotide which resulted in a new splice site.







Alternative splicing of this gene results in several transcript variants, some of







which have been characterized by the presence or absence of an ALU cassette







insert and a short or long C-terminal region. [provided by RefSeq, July 2008].







Transcript Variant: This variant (1), also known as ADAR2a-L1 or DRADA2a,







lacks the ALU cassette insert and contains the long C-terminal region, as







compared to variant 2. The resulting isoform (1), also known as hRED1-Short,







lacks an internal segment, compared to isoform 2. Publication Note: This RefSeq







record includes a subset of the publications that are available for this gene.







Please see the Gene record to access additional publications. ##RefSeq-Attributes-







START## undergoes RNA editing :: PMID: 11717408, 12045112 ##RefSeq-







Attributes-END## ##Evidence-Data-START## Transcript exon combination ::







AB194370.1, U76420.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025084, ERS025085 [ECO:0000348] ##Evidence-Data-







END##



AGBL4
intronic
84871
cytosolic
N/A
3





carboxy-







peptidase 6




APOBEC3A
exonic
200315
DNA dC−>dU-
This gene is a member of the cytidine deaminase gene family. It is one of
4





editing enzyme
seven related genes or pseudogenes found in a cluster, thought to result from gene






APOBEC-3A
duplication, on chromosome 22. Members of the cluster encode proteins that






isoform a
are structurally and functionally related to the C to U RNA-editing cytidine







deaminase APOBEC1. The protein encoded by this gene lacks the zinc binding







activity of other family members. The protein plays a role in immunity, by







restricting transmission of foreign DNA such as viruses. One mechanism of







foreign DNA restriction is deamination of foreign double-stranded DNA







cytidines to uridines, which leads to DNA degradation. However, other mecha-







nisms are also thought to be involved, as anti-viral effect is not dependent







on deaminase activity. Two transcript variants encoding different isoforms have







been found for this gene. [provided by RefSeq, July 2012]. Transcript Variant:







This variant (1) represents the longer transcript and encodes the longer isoform







(a). Publication Note: This RefSeq record includes a subset of the publications







that are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







U03891.2, BC126416.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025084 [ECO:0000348] ##Evidence-Data-







END##



APOBEC3A_B
intronic
100913187
probable
This gene is a member of the cytidine deaminase gene family. It is one of
5





DNA dC−>dU-
seven related genes or pseudogenes found in a cluster, thought to result from gene






editing
duplication, on chromosome 22. Members of the cluster encode proteins that






enzyme
are structurally and functionally related to the C to U RNA-editing cytidine






APOBEC-3A
deaminase APOBEC1. The protein encoded by this gene lacks the zinc binding







activity of other family members. The protein plays a role in immunity, by







restricting transmission of foreign DNA such as viruses. One mechanism of







foreign DNA restriction is deamination of foreign double-stranded DNA







cytidines to uridines, which leads to DNA degradation. However, other







mechanisms are also thought to be involved, as anti-viral effect is not dependent







on deaminase activity. The protein encoded by this gene is the same as that







encoded by APOBEC3A; however, this gene is a hybrid gene that results from







the deletion of approximately 29.5 kb of sequence between the APOBEC3A gene







and the adjacent gene APOBEC3B. The breakpoints of the deletion are within







the two genes, so the deletion hybrid is predicted to have the promoter and







coding region of APOBEC3A, but the 3′ UTR of APOBEC3B. [provided by







RefSeq, July 2012]. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications. ##Evidence-Data-START## RNAseq introns :: single







sample supports all introns ERS025081, ERS025084 [ECO:0000348]







##Evidence-Data-END##



APOBEC3B
exonic
9582
DNA dC−>dU-
This gene is a member of the cytidine deaminase gene family. It is one of seven
6





editing enzyme
related genes or pseudogenes found in a cluster, thought to result from gene






APOBEC-3B
duplication, on chromosome 22. Members of the cluster encode proteins that are






isoform a
structurally and functionally related to the C to U RNA-editing cytidine







deaminase APOBEC1. It is thought that the proteins may be RNA editing







enzymes and have roles in growth or cell cycle control. A hybrid gene results







from the deletion of approximately 29.5 kb of sequence between this gene,







APOBEC3B, and the adjacent gene APOBEC3A. The breakpoints of the deletion







are within the two genes, so the deletion allele is predicted to have the promoter







and coding region of APOBEC3A, but the 3′ UTR of APOBEC3B. Two







transcript variants encoding different isoforms have been found for this gene.







[provided by RefSeq, July 2012]. Transcript Variant: This variant (1) represents







the longer transcript and encodes the longer isoform (a). Publication Note: This







RefSeq record includes a subset of the publications that are available for this







gene. Please see the Gene record to access additional publications. ##Evidence-







Data-START## Transcript exon combination :: AY743217.1 [ECO:0000332]







RNAseq introns :: mixed/partial sample support ERS025081, ERS025082







[ECO:0000350] ##Evidence-Data-END##



APOLD1
exonic
81575
apolipoprotein L
APOLD1 is an endothelial cell early response protein that may play a role in
7





domain-
regulation of endothelial cell signaling and vascular function (Regard et al., 2004






containing
[PubMed 15102925]). [supplied by OMIM, December 2008]. Transcript






protein 1
Variant: This variant (1) represents the longer transcript and encodes the longer






isoform 1
isoform (1). Sequence Note: This RefSeq record was created from transcript and







genomic sequence data to make the sequence consistent with the reference







genome assembly. The extent of this transcript is supported by transcript







alignments. ##Evidence-Data-START## Transcript exon combination ::







BC042478.1, DR000985.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025086 [ECO:0000348] ##Evidence-







Data-END##



ARHGEF7
exonic
8874
rho guanine
Rho GTPases play a fundamental role in numerous cellular processes triggered
8





nucleotide
by extracellular stimuli that work through G protein coupled receptors. The






exchange
encoded protein belongs to a family of cytoplasmic proteins that activate the Ras-






factor 7
like family of Rho proteins by exchanging bound GDP for GTP. It forms a






isoform a
complex with the small GTP binding protein Rac1 and recruits Rac1 to







membrane ruffles and to focal adhesions. This protein can induce membrane







ruffling Multiple alternatively spliced transcript variants encoding different







isoforms have been described for this gene. [provided by RefSeq, July 2008].







Transcript Variant: This variant (1) differs in the 5′ UTR, 3′ UTR, coding region,







and uses a downstream start codon, compared to variant 3. Both variants 1 and 5







encode isoform a, which has a shorter N-terminus and a longer and distinct C-







terminus, compared to isoform c. Publication Note: This RefSeq record includes







a subset of the publications that are available for this gene. Please see the Gene







record to access additional publications. ##Evidence-Data-START## Transcript







exon combination :: D63476.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-







END##



ASTN2
intronic
23245
astrotactin-2
This gene encodes a protein that is expressed in the brain and may function in
9





isoform a
neuronal migration, based on functional studies of the related astrotactin 1 gene






precursor
in human and mouse. A deletion at this locus has been associated with







schizophrenia. Multiple transcript variants encoding different proteins have been







found for this locus. [provided by RefSeq, May 2010]. Transcript Variant: This







variant (1) represents the longest transcript and encodes the longest isoform (a).







##Evidence-Data-START## Transcript exon combination :: BC146756.1,







AB014534.1 [ECO:0000332] RNAseq introns :: single sample supports all







introns ERS025082 [ECO:0000348] ##Evidence-Data-END##



AUH
exonic
549
methylglutaconyl-
The methylglutaconyl-CoA hydratase, mitochondrial protein binds to the AU-
10





CoA hydratase,
rich element (ARE), a common element found in the 3′ UTR of rapidly decaying






mitochondrial
mRNA such as c-fos, c-myc and granulocyte/macrophage colony stimulating






precursor
factor. ARE elements are involved in directing RNA to rapid degradation and







deadenylation. AUH is also homologous to enol-CoA hydratase, an enzyme







involved in fatty acid degradation, and has been shown to have intrinsic







hydratase enzymatic activity. AUH is thus a bifunctional chimera between RNA







binding and metabolic enzyme activity. A possible subcellular localization in the







mitochondria has been demonstrated for the mouse homolog of this protein







which shares 92% identity with the human protein. It has been suggested that







AUH may have a novel role as a mitochondrial located AU-binding protein.







Human AUH is expressed as a single mRNA species of 1.8 kb, and translated as







a 40-kDa precursor protein which is subsequently processed to a 32-kDa mature







form. [provided by RefSeq, May 2010]. Publication Note: This RefSeq record







includes a subset of the publications that are available for this gene. Please see







the Gene record to access additional publications. ##RefSeq-Attributes-







START## gene product(s) localized to mito. :: reported by MitoCarta ##RefSeq-







Attributes-END## ##Evidence-Data-START## Transcript exon combination ::







X79888.1, AL533438.3 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025084, ERS025088 [ECO:0000348] ##Evidence-Data-







END##



BACH1
exonic
571
BTB Domain
This gene encodes a transcription factor that belongs to the cap‘n’collar type of
11





And CNC
basic region leucine zipper factor family (CNC-bZip). The encoded protein






Homolog 1
contains broad complex, tramtrack, bric-a-brac/poxvirus and zinc finger







(BTB/POZ) domains, which is atypical of CNC-bZip family members. These







BTB/POZ domains facilitate protein-protein interactions and formation of homo-







and/or hetero-oligomers. When this encoded protein forms a heterodimer with







MafK, it functions as a repressor of Maf recognition element (MARE) and







transcription is repressed. Multiple alternatively spliced transcript variants have







been identified for this gene. [provided by RefSeq, May 2009]. Transcript







Variant: This variant (3), also named BACH1t, differs in the 5′ UTR, 3′ coding







region and 3′ UTR (compared to variant 1). This variant is represented as non-







coding because the use of the 5′-most supported translational start codon, as used







in variant 1, renders the transcript a candidate for nonsense-mediated mRNA







decay (NMD). This transcript represents the splice variant reported by Kanezaki







et al. (PMID: 11069897). Publication Note: This RefSeq record includes a subset







of the publications that are available for this gene. Please see the Gene record to







access additional publications. ##Evidence-Data-START## RNAseq introns ::







mixed/partial sample support ERS025084, ERS025088 [ECO:0000350]







##Evidence-Data-END##



BDKRB2
intronic
624
B2 bradykinin
This gene encodes a receptor for bradykinin. The 9 aa bradykinin peptide elicits
12





receptor
many responses including vasodilation, edema, smooth muscle spasm and pain







fiber stimulation. This receptor associates with G proteins that stimulate a







phosphatidylinositol-calcium second messenger system. Alternate start codons







result in two isoforms of the protein. [provided by RefSeq, July 2008].







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







DC369062.1, DC417219.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025090 [ECO:0000348] ##Evidence-Data-







END##



BMPR2
intronic
659
bone
This gene encodes a member of the bone morphogenetic protein (BMP) receptor
13





morphogenetic
family of transmembrane serine/threonine kinases. The ligands of this receptor






protein receptor
are BMPs, which are members of the TGF-beta superfamily. BMPs are involved






type-2 precursor
in endochondral bone formation and embryogenesis. These proteins transduce







their signals through the formation of heteromeric complexes of two different







types of serine (threonine) kinase receptors: type I receptors of about 50-55 kD







and type II receptors of about 70-80 kD. Type II receptors bind ligands in the







absence of type I receptors, but they require their respective type I receptors for







signaling, whereas type I receptors require their respective type II receptors for







ligand binding. Mutations in this gene have been associated with primary







pulmonary hypertension, both familial and fenfluramine-associated, and with







pulmonary venoocclusive disease. [provided by RefSeq, July 2008]. Sequence







Note: This RefSeq record was created from transcript and genomic sequence data







to make the sequence consistent with the reference genome assembly. The extent







of this transcript is supported by transcript alignments. Publication Note: This







RefSeq record includes a subset of the publications that are available for this







gene. Please see the Gene record to access additional publications. ##Evidence-







Data-START## Transcript exon combination :: BC052985.2, AK292430.1







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-END##



BTBD17
exonic
388419
BTB/POZ
N/A
14





domain-







containing







protein 17







precursor




C17orf77
exonic
146723
uncharacterized
N/A
15





protein







C17orf77







precursor




CAPZB
intronic
832
F-actin-
This gene encodes the beta subunit of the barbed-end actin binding protein,
16





capping protein
which belongs to the F-actin capping protein family. The capping protein is a






subunit beta
heterodimeric actin capping protein that blocks actin filament assembly and






isoform 1
disassembly at the fast growing (barbed) filament ends and functions in







regulating actin filament dynamics as well as in stabilizing actin filament lengths







in muscle and nonmuscle cells. A pseudogene of this gene is located on the long







arm of chromosome 2. Multiple alternatively spliced transcript variants encoding







different isoforms have been found. [provided by RefSeq, August 2013].







Transcript Variant: This variant (1) encodes isoform 1. Publication Note: This







RefSeq record includes a subset of the publications that are available for this







gene. Please see the Gene record to access additional publications.







##Evidence-Data-START## Transcript exon combination :: BC107752.1,







BM451686.1 [ECO:0000332] RNAseq introns :: single sample supports







all introns ERS025088 [ECO:0000348] ##Evidence-Data-END##



CCDC41
exonic
51134
centrosomal
N/A
17





protein of 83 kDa




CD300A
exonic
11314
CMRF35-like
This gene encodes a member of the CD300 glycoprotein family of cell surface
18





molecule 8
proteins found on leukocytes involved in immune response signaling pathways.






isoform 1
This gene is located on chromosome 17 in a cluster with all but one of the other






precursor
family members. Multiple transcript variants encoding different isoforms have







been found for this gene. [provided by RefSeq, February 2012]. Transcript







Variant: This variant (1) represents the longer transcript and encodes the longer







protein (isoform 1), also referred to as IRC1a. Publication Note: This RefSeq







record includes a subset of the publications that are available for this gene.







Please see the Gene record to access additional publications. ##Evidence-







Data-START## Transcript exon combination :: BC032352.1, AL531420.3







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025081, ERS025083 [ECO:0000348] ##Evidence-Data-END##



CD300C
exonic
10871
CMRF35-like
The CMRF35 antigen, which was identified by reactivity with a monoclonal
19





molecule 6
antibody, is present on monocytes, neutrophils, and some T and B lymphocytes






precursor
(Jackson et al., 1992 [PubMed 1349532]). [supplied by OMIM, March 2008].







##Evidence-Data-START## Transcript exon combination :: BC022279.1,







BM922826.1 [ECO:0000332] RNAseq introns :: single sample supports all







introns ERS025084, ERS025087 [ECO:0000348] ##Evidence-Data-END##



CD300E
exonic
342510
CMRF35-like
This gene encodes a member of the CD300 glycoprotein family of cell surface
20





molecule 2
proteins expressed on myeloid cells. The protein interacts with the TYRO protein






precursor
tyrosine kinase-binding protein and is thought to act as an activating receptor.







[provided by RefSeq, November 2012]. Sequence Note: This RefSeq record was







created from transcript and genomic sequence data to make the sequence







consistent with the reference genome assembly. The genomic coordinates used







for the transcript record were based on transcript alignments. An in-frame AUG







is located 41 codons upstream of the annotated translation start site but is not







being annotated as a start site since it is not conserved and is in a weak Kozak







sequence context. ##RefSeq-Attributes-START## CDS uses downstream in-







frame AUG :: downstream AUG is associated with N-terminal localization signal







##RefSeq-Attributes-END## ##Evidence-Data-START## Transcript exon







combination :: AK303545.1, BX648376.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025084, ERS025088 [ECO:0000348]







##Evidence-Data-END##



CD300LB
exonic
124599
CMRF35-like
CD300LB is a nonclassical activating receptor of the immunoglobulin (Ig)
21





molecule 7
superfamily expressed on myeloid cells (Martinez-Barriocanal and Sayos, 2006






precursor
[PubMed 16920917]).[supplied by OMIM, March 2008]. CCDS Note: The coding







region has been updated to shorten the N-terminus to one that is more supported







by available conservation data and paralogous family members. The update has a







predicted N-terminal signal peptide, which is consistent with functional support







for the protein (e.g., PMIDs 16920917, 19359216). ##Evidence-Data-START##







Transcript exon combination :: BC028091.1, AY359025.1 [ECO:0000332]







RNAseq introns :: single sample supports all introns ERS025084, ERS025088







[ECO:0000348] ##Evidence-Data-END## ##RefSeq-Attributes-START## CDS







uses downstream in-frame AUG :: downstream AUG is associated with N-







terminal localization signal ##RefSeq-Attributes-END##



CD300LD
exonic
100131439
CMRF35-like
N/A
22





molecule 4







precursor




CD300LF
exonic
146722
CMRF35-like
CD300LF is an inhibitory receptor of the Ig superfamily expressed on myeloid
23





molecule 1
cells. It mediates negative regulatory signals by recruiting SHP1 (PTPN6; MIM






precursor
176883) or SHIP (INPP5D; MIM 601582) (Sui et al., 2004 [PubMed 15184070];







Alvarez-Errico et al., 2004 [PubMed 15549731]). [supplied by OMIM, March







2008]. Sequence Note: The RefSeq transcript and protein were derived from







genomic sequence to make the sequence consistent with the reference genome







assembly. The genomic coordinates used for the transcript record were based on







alignments. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications. ##Evidence-Data-START## Transcript exon







combination :: AF251706.1, AY358545.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025084 [ECO:0000348] ##Evidence-







Data-END##



CDKN1B
exonic
1027
cyclin-dependent
This gene encodes a cyclin-dependent kinase inhibitor, which shares a limited
24





kinase inhibitor
similarity with CDK inhibitor CDKN1A/p21. The encoded protein binds to and






1B
prevents the activation of cyclin E-CDK2 or cyclin D-CDK4 complexes, and







thus controls the cell cycle progression at G1. The degradation of this protein,







which is triggered by its CDK dependent phosphorylation and subsequent







ubiquitination by SCF complexes, is required for the cellular transition from







quiescence to the proliferative state. [provided by RefSeq, July 2008]. Publication







Note: This RefSeq record includes a subset of the publications that are available







for this gene. Please see the Gene record to access additional publications.







##Evidence-Data-START## Transcript exon combination :: BC001971.1,







AY004255.1 [ECO:0000332] RNAseq introns :: single sample supports all







introns ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-END##



CENPM
exonic
79019
centromere
The centromere is a specialized chromatin domain, present throughout the cell
25





protein
cycle, that acts as a platform on which the transient assembly of the kinetochore






M isoform a
occurs during mitosis. All active centromeres are characterized by the presence







of long arrays of nucleosomes in which CENPA (MIM 117139) replaces histone







H3 (see MIM 601128). CENPM is an additional factor required for centromere







assembly (Foltz et al., 2006 [PubMed 16622419]). [supplied by OMIM, March







2008]. Transcript Variant: This variant (1) represents the longer transcript and







encodes the longer isoform (a). Publication Note: This RefSeq record includes a







subset of the publications that are available for this gene. Please see the Gene







record to access additional publications. ##Evidence-Data-START## Transcript







exon combination :: BC000705.2, BC007495.2 [ECO:0000332] RNAseq







introns :: single sample supports all introns ERS025085, ERS025088







[ECO:0000348] ##Evidence-Data-END##



COG4
exonic
25839
conserved
The protein encoded by this gene is a component of an oligomeric protein
26





oligomeric Golgi
complex involved in the structure and function of the Golgi apparatus. Defects in






complex subunit
this gene may be a cause of congenital disorder of glycosylation type IIj. Two






4 isoform 1
transcript variants encoding different isoforms have been found for this







gene. [provided by RefSeq, August 2010]. Transcript Variant: This variant (1)







represents the longer transcript and encodes the longer isoform (1). ##Evidence-







Data-START## Transcript exon combination :: BC072438.1, AK022874.1







[ECO:0000332] RNAseq introns :: mixed/partial sample support ERS025081,







ERS025082 [ECO:0000350] ##Evidence-Data-END##



COMMD6
exonic
170622
COMM domain-
COMMD6 belongs to a family of NF-kappa-B (see RELA; MIM 164014)-
27





containing
inhibiting proteins characterized by the presence of a COMM domain (see






protein
COMMD1; MIM 607238) (de Bie et al., 2006 [PubMed 16573520]). [supplied by






6 isoform a
OMIM, March 2009]. ##Evidence-Data-START## Transcript exon combination ::







HY028175.1, DW440523.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025088 [ECO:0000348] ##Evidence-Data-END##



CRADD
exonic
8738
death domain-
The protein encoded by this gene is a death domain (CARD/DD)-containing
28





containing
protein and has been shown to induce cell apoptosis. Through its CARD domain,






protein
this protein interacts with, and thus recruits, caspase 2/ICH1 to the cell death






CRADD
signal transduction complex that includes tumor necrosis factor receptor 1







(TNFR1A), RIPK1/RIP kinase, and numbers of other CARD domain-containing







proteins. [provided by RefSeq, July 2008]. Publication Note: This RefSeq record







includes a subset of the publications that are available for this gene. Please see







the Gene record to access additional publications. ##Evidence-Data-START##







Transcript exon combination :: BX480215.1, BC017042.1 [ECO:0000332]







RNAseq introns :: single sample supports all introns ERS025081, ERS025083







[ECO:0000348] ##Evidence-Data-END##



CREBL2
exonic
1389
cAMP-responsive
cAMP response element (CRE)-binding protein-like-2 (CREBL2) was identified
29





element-binding
in a search to find genes in a commonly deleted region on chromosome 12p13






protein-like 2
flanked by ETV6 and CDKN1B genes, frequently associated with hematopoietic







malignancies, as well as breast, non-small-cell lung and ovarian cancers.







CREBL2 shares a 41% identity with CRE-binding protein (CREB) over a 48-







base long region which encodes the bZip domain of CREB. The bZip domain







consists of about 30 amino acids rich in basic residues involved in DNA binding,







followed by a leucine zipper motif involved in protein dimerization. This







suggests that CREBL2 encodes a protein with DNA binding capabilities. The







occurance of CREBL2 deletion in malignancy suggests that CREBL2 may act as







a tumor suppressor gene. [provided by RefSeq, July 2008]. ##Evidence-Data-







START## Transcript exon combination :: BC106052.1, AF039081.1







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-END##



DNAI2
exonic
64446
dynein
The protein encoded by this gene belongs to the dynein intermediate chain
30





intermediate
family, and is part of the dynein complex of respiratory cilia and sperm flagella






chain 2,
Mutations in this gene are associated with primary ciliary dyskinesia type 9.






axonemal
Alternatively spliced transcript variants encoding different isoforms have been






isoform 1
noted for this gene. [provided by RefSeq, March 2010]. Transcript Variant: This







variant (1) encodes the longer isoform (1). ##Evidence-Data-START##







Transcript exon combination :: AF250288.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025085 [ECO:0000348] ##Evidence-







Data-END## ##RefSeq-Attributes-START## NMD candidate :: translation







inferred from conservation ##RefSeq-Attributes-END##



DNER
intronic
92737
delta and Notch-
N/A
31





like epidermal







growth factor-







related receptor







precursor




DUSP16
exonic
80824
dual specificity
This gene encodes a mitogen-activated protein kinase phosphatase that is a
32





protein
member of the dual specificity protein phosphatase subfamily. These






phosphatase 16
phosphatases inactivate their target kinases by dephosphorylating both the







phosphoserine/threonine and phosphotyrosine residues. The encoded protein







specifically regulates the c-Jun amino-terminal kinase (JNK) and extracellular







signal-regulated kinase (ERK) pathways. [provided by RefSeq, May 2010].







Sequence Note: This RefSeq record was created from transcript and genomic







sequence data to make the sequence consistent with the reference genome







assembly. The genomic coordinates used for the transcript record were based on







transcript alignments. Publication Note: This RefSeq record includes a subset of







the publications that are available for this gene. Please see the Gene record to







access additional publications. ##Evidence-Data-START## Transcript exon







combination :: AF506796.1, AB052156.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025084, ERS025088 [ECO:0000348]







##Evidence-Data-END##



ECRP
exonic
643332
N/A
N/A
33


EDIL3
intronic
10085
EGF-like repeat
The protein encoded by this gene is an integrin ligand. It plays an important role
34





and discoidin I-
in mediating angiogenesis and may be important in vessel wall remodeling and






like domain-
development. It also influences endothelial cell behavior. [provided by RefSeq,






containing
July 2008]. Transcript Variant: This variant (1) encodes the longer isoform (1).






protein 3
Sequence Note: This RefSeq record was created from transcript and genomic






isoform
sequence data to make the sequence consistent with the reference genome






1 precursor
assembly. The genomic coordinates used for the transcript record were based on







transcript alignments. Publication Note: This RefSeq record includes a subset of







the publications that are available for this gene. Please see the Gene record to







access additional publications. ##Evidence-Data-START## Transcript exon







combination :: BC030828.1, U70312.1 [ECO:0000332] RNAseq introns ::







mixed/partial sample support ERS025081, ERS025082 [ECO:0000350]







##Evidence-Data-END##



EEA1
exonic
8411
early endosome
N/A
35





antigen 1




EHF
both
26298
ETS homologous
This gene encodes a protein that belongs to an ETS transcription factor
36





factor isoform 1
subfamily characterized by epithelial-specific expression (ESEs). The encoded






precursor
protein acts as a transcriptional repressor and may be involved in epithelial







differentiation and carcinogenesis. Three transcript variants encoding different







isoforms have been found for this gene. [provided by RefSeq, June 2011].







Transcript Variant: This variant (1) encodes the longest isoform (1). Sequence







Note: This RefSeq record was created from transcript and genomic sequence data







to make the sequence consistent with the reference genome assembly. The







genomic coordinates used for the transcript record were based on transcript







alignments. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications. ##Evidence-Data-START## Transcript exon







combination :: AK310867.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025084, ERS025086 [ECO:0000348] ##Evidence-Data-







END##



EMB
exonic
133418
embigin precursor
This gene encodes a transmembrane glycoprotein that is a member of the
37






immunoglobulin superfamily. The encoded protein may be involved in cell







growth and development by mediating interactions between the cell and







extracellular matrix. A pseudogene of this gene is found on chromosome 1.







[provided by RefSeq, January 2009]. ##Evidence-Data-START## Transcript exon







combination :: BC059398.1, AK300860.1 [ECO:0000332] RNAseq introns ::







mixed/partial sample support ERS025081, ERS025082 [ECO:0000350]







##Evidence-Data-END##



ETV6
exonic
2120
transcription
This gene encodes an ETS family transcription factor. The product of this gene
38





factor
contains two functional domains: a N-terminal pointed (PNT) domain that is






ETV6
involved in protein-protein interactions with itself and other proteins, and a C-







terminal DNA-binding domain. Gene knockout studies in mice suggest that it is







required for hematopoiesis and maintenance of the developing vascular network.







This gene is known to be involved in a large number of chromosomal







rearrangements associated with leukemia and congenital fibrosarcoma. [provided







by RefSeq, September 2008]. Publication Note: This RefSeq record includes a







subset of the publications that are available for this gene. Please see the Gene







record to access additional publications. ##Evidence-Data-START##







Transcript exon combination :: BC043399.1, U11732.1 [ECO:0000332] RNAseq







introns :: singlesample supports all introns ERS025081, ERS025082







[ECO:0000348] ##Evidence-Data-END##



FHL2
exonic
2274
four and a
This gene encodes a member of the four-and-a-half-LIM-only protein family.
39





half LIM
Family members contain two highly conserved, tandemly arranged, zinc finger






domains
domains with four highly conserved cysteines binding a zinc atom in each zinc






protein 2
finger. This protein is thought to have a role in the assembly of extracellular







membranes. Also, this gene is down-regulated during transformation of normal







myoblasts to rhabdomyosarcoma cells and the encoded protein may function as a







link between presenilin-2 and an intracellular signaling pathway. Multiple







alternatively spliced variants, encoding the same protein, have been identified.







[provided by RefSeq, August 2011]. Transcript Variant: This variant (1) differs in







the 5′ UTR compared to variant 2. Variants 1, 2, 4 and 5 encode the same







isoform. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications. ##RefSeq-Attributes-START## CDS uses downstream







in-frame AUG :: lack of evidence for use of upstream AUG ##RefSeq-







Attributes-END## ##Evidence-Data-START## Transcript exon combination ::







BC093049.1, AL523628.3 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025084, ERS025088 [ECO:0000348] ##Evidence-Data-







END##



FLJ26850
intronic
400710
N/A
N/A
40


FPR2
exonic
2358
N-formyl peptide
N/A
41





receptor 2




FPR3
exonic
2359
N-formyl peptide
N/A
42





receptor 3




FUK
both
197258
L-fucose kinase
The protein encoded by this gene belongs to the GHMP (galacto-, homoserine,
43






mevalonate and phosphomevalonate) kinase family and catalyzes the







phosphorylation of L-fucose to form beta-L-fucose 1-phosphate. This enzyme







catalyzes the first step in the utilization of free L-fucose in glycoprotein and







glycolipid synthesis. L-fucose may be important in mediating a number of cell-







cell interactions such as blood group antigen recognition, inflammation, and







metastatis. While several transcript variants may exist for this gene, the full-







length nature of only one has been described to date. [provided by RefSeq, July







2008]. ##Evidence-Data-START## Transcript exon combination :: AJ441184.1,







BC032542.1 [ECO:0000332] RNAseq introns :: mixed/partial sample support







ERS025081, ERS025082 [ECO:0000350] ##Evidence-Data-END##



GDA
exonic
9615
guanine
This gene encodes an enzyme responsible for the hydrolytic deamination of
44





deaminase
guanine. Studies in rat ortholog suggest this gene plays a role in microtubule






isoform a
assembly. Multiple transcript variants encoding different isoforms have been







found for this gene. [provided by RefSeq, November 2011]. Transcript Variant:







This variant (1) encodes the longest isoform (a). Sequence Note: This RefSeq







record was created from transcript and genomic sequence data to make the







sequence consistent with the reference genome assembly. The genomic







coordinates used for the transcript record were based on transcript alignments.







##Evidence-Data-START## RNAseq introns :: mixed/partial sample







support ERS025082, ERS025083 [ECO:0000350] ##Evidence-Data-END##



GDPD4
exonic
220032
glycerophos-
N/A
45





phodiester







phosphodiesterase







domain-







containing







protein 4




GPATCH2
intronic
55105
G patch domain-
N/A
46





containing







protein 2




GPC5
intronic
2262
glypican-5
Cell surface heparan sulfate proteoglycans are composed of a membrane-
47





precursor
associated protein core substituted with a variable number of heparan sulfate







chains. Members of the glypican-related integral membrane proteoglycan family







(GRIPS) contain a core protein anchored to the cytoplasmic membrane via a







glycosyl phosphatidylinositol linkage. These proteins may play a role in the







control of cell division and growth regulation. [provided by RefSeq, July 2008].







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







BC030584.1, BC039730.1 [ECO:0000332] RNAseq introns :: mixed/partial







sample support ERS025082, ERS025083 [ECO:0000350] ##Evidence-Data-







END##



GPR19
exonic
2842
probable
N/A
48





G-protein coupled







receptor 19




GPR142
exonic
350383
probable
GPR142 is a member of the rhodopsin family of G protein-coupled receptors
49





G-protein
(GPRs) (Fredriksson et al., 2003 [PubMed 14623098]). [supplied by OMIM,






coupled
March 2008]. ##Evidence-Data-START## Transcript exon combination ::






receptor 142
AB196530.1, AY288421.1 [ECO:0000332] ##Evidence-Data-END##



GPRC5C
exonic
55890
G-protein coupled
The protein encoded by this gene is a member of the type 3 G protein-coupled
50





receptor family C
receptor family. Members of this supeifamily are characterized by a signature 7-






groups 5 member
transmembrane domain motif. The specific function of this protein is unknown;






C isoform a
however, this protein may mediate the cellular effects of retinoic acid on the G







protein signal transduction cascade. Two transcript variants encoding different







isoforms have been found for this gene. [provided by RefSeq, July 2008].







Transcript Variant: This variant (1) represents the longer transcript and encodes







the longer isoform (a). ##Evidence-Data-START## Transcript exon







combination :: BC110848.1, AK131210.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025081, ERS025083 [ECO:0000348]







##Evidence-Data-END##



GRIA3
intronic
2892
glutamate
Glutamate receptors are the predominant excitatory neurotransmitter receptors in
51





receptor 3
the mammalian brain and are activated in a variety of normal neurophysiologic






isoform 1
processes. These receptors are heteromeric protein complexes composed of






precursor
multiple subunits, arranged to form ligand-gated ion channels. The classification







of glutamate receptors is based on their activation by different pharmacologic







agonists. The subunit encoded by this gene belongs to a family of AMPA (alpha-







amino-3-hydroxy-5-methyl-4-isoxazole propionate)-sensitive glutamate







receptors, and is subject to RNA editing.(AGA−>GGA; R−>G). Alternative .







splicing at this locus results in different isoforms, which may vary in their signal







transduction properties. [provided by RefSeq, July 2008]. Transcript Variant: This







variant (1) encodes isoform 1 (also known as flip isoform). RNA editing (AGA−>







GGA) changes Arg775Gly. Publication Note: This RefSeq record includes a







subset of the publications that are available for this gene. Please see the Gene







record to access additional publications. ##RefSeq-Attributes-START##







undergoes RNA editing :: PMID: 10688364, 7992055 ##RefSeq-Attributes-







END## ##Evidence-Data-START## Transcript exon combination :: U10301.1







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025082, ERS025084 [ECO:0000348] ##Evidence-Data-END##



GTPBP4
exonic
23560
nucleolar GTP-
GTP-binding proteins are GTPases and function as molecular switches that can
52





binding
flip between two states: active, when GTP is bound, and inactive, when GDP is






protein 1
bound. ‘Active’ in this context usually means that the molecule acts as a signal to







trigger other events in the cell. When an extracellular ligand binds to a G-protein-







linked receptor, the receptor changes its conformation and switches on the







trimeric G proteins that associate with it by causing them to eject their GDP and







replace it with GTP. The switch is turned off when the G protein hydrolyzes its







own bound GTP, converting it back to GDP. But before that occurs, the active







protein has an opportunity to diffuse away from the receptor and deliver its







message for a prolonged period to its downstream target. [provided by RefSeq,







July 2008]. ##Evidence-Data-START## Transcript exon combination ::







AK001552.1, AK222861.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-







END##



HCN1
exonic
348980
potassium/sodium
The membrane protein encoded by this gene is a hyperpolarization-activated
53





hyperpolarization-
cation channel that contributes to the native pacemaker currents in heart and






activated cyclic
neurons. The encoded protein can homodimerize or heterodimerize with other






nucleotide-gated
pore-forming subunits to form a potassium channel. This channel may act as a






channel 1
receptor for sour tastes. [provided by RefSeq, October 2011]. Sequence Note:







This RefSeq record was created from transcript and genomic sequence data to







make the sequence consistent with the reference genome assembly. The genomic







coordinates used for the transcript record were based on transcript alignments.







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







AF488549.1, AF064876.1 [ECO:0000332] RNAseq introns :: mixed/partial







sample support ERS025081, ERS025082 [ECO:0000350] ##Evidence-Data-







END##



HEXA
exonic
3073
beta-
This gene encodes the alpha subunit of the lysosomal enzyme beta-
54





hexosaminidase
hexosainidase that, together with the cofactor GM2 activator protein, catalyzes






subunit alpha
the degradation of the ganglioside GM2, and other molecules containing terminal






preproprotein
N-acetyl hexosamines. Beta-hexosaminidase is composed of two subunits, alpha







and beta, which are encoded by separate genes. Bothbeta-hexosaminidase alpha







and beta subunits are members of family 20 of glycosyl hydrolases. Mutations in







the alpha or beta subunit genes lead to an accumulation of GM2 ganglioside in







neurons and neurodegenerative disorders termed the GM2 gangliosidoses. Alpha







subunit gene mutations lead to Tay-Sachs disease (GM2-gangliosidosis type I).







[provided by RefSeq, July 2009]. Sequence Note: This RefSeq record was created







from transcript and genomic sequence data because no single transcript was







available for the full length of the gene. The extent of this transcript is supported







by transcript alignments. Publication Note: This RefSeq record includes a subset







of the publications that are available for this gene. Please see the Gene record to







access additional publications. ##Evidence-Data-START## Transcript exon







combination :: M13520.1, CR627386.1 [ECO:0000332] RNAseq introns :: single







sample supports all introns ERS025084, ERS025088 [ECO:0000348]







##Evidence-Data-END##



HK2
exonic
3099
hexokinase-2
Hexokinases phosphorylate glucose to produce glucose-6-phosphate, the first
55






step in most glucose metabolism pathways. This gene encodes hexokinase 2, the







predominant form found in skeletal muscle. It localizes to the outer membrane of







mitochondria. Expression of this gene is insulin-responsive, and studies in rat







suggest that it is involved in the increased rate of glycolysis seen in rapidly







growing cancer cells. [provided by RefSeq, April 2009]. Publication Note: This







RefSeq record includes a subset of the publications that are available for this







gene. Please see the Gene record to access additional publications. ##Evidence-







Data-START## Transcript exon combination :: BC064369.1, AF148513.1







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025083, ERS025084 [ECO:0000348] ##Evidence-Data-END##



HMGB3
exonic
3149
high mobility
HMGB3 belongs to the high mobility group (HMG) protein superfamily. Like
56





group protein B3
HMG1 (MIM 163905) and HMG2 (MIM 163906), HMGB3 contains DNA-







binding HMG box domains and is classified into the HMG box subfamily.







Members of the HMG box subfamily are thought to play a fundamental role in







DNA replication, nucleosome assembly and transcription (Wilke et al., 1997







[PubMed 9370291]; Nemeth et al., 2006 [PubMed 16945912]). [supplied by







OMIM, Mar. 2008]. ##Evidence-Data-START## Transcript exon combination ::







Y10043.1, BG176733.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-







END##



HPR
exonic
3250
haptoglobin-
This gene encodes a haptoglobin-related protein that binds hemoglobin as
57





related
efficiently as haptoglobin. Unlike haptoglobin, plasma concentration of this






protein
protein is unaffected in patients with sickle cell anemia and extensive






precursor
intravascular hemolysis, suggesting a difference in binding between haptoglobin-







hemoglobin and haptoglobin-related protein-hemoglobin complexes to CD163,







the hemoglobin scavenger receptor. This protein may also be a clinically







important predictor of recurrence of breast cancer. [provided by RefSeq, October







2011]. Publication Note: This RefSeq record includes a subset of the publications







that are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







CB147217.1, CB122261.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025084, ERS025088 [ECO:0000348] ##Evidence-Data-







END##



HTATSF1P2
exonic
401233
N/A
N/A
58


IDI2
exonic
91734
isopentenyl-
N/A
59





diphosphate







Delta-







isomerase 2




IDI2-AS1
exonic
55853
N/A
N/A
60


IDO2
intronic
169355
indoleamine 2,3-
Along with the enzymes encoded by the INDO (MIM 147435) and TDO2 (MIM
61





dioxygenase 2
191070) genes, the enzyme encoded by the INDOL1 gene metabolizes







tryptophan in the kynurenine pathway (Ball et al., 2007 [PubMed







17499941]). [supplied by OMIM, February 2011]. Sequence Note: The RefSeq







transcript 3′ UTR was derived from genomic sequence to make the sequence







consistent with the reference genome assembly. The genomic coordinates used







were based on transcript alignments.



IFNLR1
exonic
163702
interferon
The protein encoded by this gene belongs to the class II cytokine receptor family.
62





lambda
This protein forms a receptor complex with interleukine 10 receptor, beta






receptor
(IL10RB). The receptor complex has been shown to interact with three closely






1 isoform
related cytokines, including interleukin 28A (IL28A), interleukin 28B (IL28B),






1 precursor
and interleukin 29 (IL29). The expression of all three cytokines can be induced







by viral infection. The cells overexpressing this protein have been found to have







enhanced responses to IL28A and IL29, but decreased response to IL28B. Three







alternatively spliced transcript variants encoding distinct isoforms have been







reported. [provided by RefSeq, July 2008]. Transcript Variant: This variant (1)







represents the longest transcript and it encodes the longest protein (isoform 1).







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







AF439325.1, AK160364.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025084 [ECO:0000348] ##Evidence-Data-END##



IQCB1
exonic
9657
IQ calmodulin-
This gene encodes a nephrocystin protein that interacts with calmodulin and the
63





binding motif-
retinitis pigmentosa GTPase regulator protein. The encoded protein has a central






containing
coiled-coil region and two calmodulin-binding IQ domains. It is localized to the






protein 1
primary cilia of renal epithelial cells and connecting cilia of photoreceptor cells.






isoform a
The protein is thought to play a role in ciliary function. Defects in this gene result







in Senior-Loken syndrome type 5. Alternative splicing results in multiple







transcript variants. [provided by RefSeq, November 2009]. Transcript Variant:







This variant (1) encodes the longer isoform (a). Publication Note: This RefSeq







record includes a subset of the publications that are available for this gene.







Please see the Gene record to access additional publications. ##Evidence-







Data-START## Transcript exon combination :: D25278.1, AY714228.1







[ECO:0000332] RNAseq introns :: mixed/partial sample support ERS025081,







ERS025082 [ECO:0000350] ##Evidence-Data-END##



JPX
intronic
554203

JPX is a nonprotein-coding RNA transcribed from a gene within the X-
64






inactivation center (XIC; MIM 314670) that appears to participate in X







chromosome inactivation (Tian et al., 2010 [PubMed 21029862]). [supplied by







OMIM, February 2011]. ##Evidence-Data-START## Transcript exon







combination :: BC071776.1 [ECO:0000332] RNAseq introns :: mixed/partial







sample support ERS025081, ERS025082 [ECO:0000350] ##Evidence-







Data-END##



KANK1
intronic
23189
KN motif and
The protein encoded by this gene belongs to the Kank family of proteins, which
65





ankyrin repeat
contain multiple ankyrin repeat domains. This family member functions in






domain-
cytoskeleton formation by regulating actin polymerization. This gene is a






containing
candidate tumor suppressor for renal cell carcinoma. Mutations in this gene cause






protein 1
cerebral palsy spastic quadriplegic type 2, a central nervous system development






isoform a
disorder. A t(5; 9) translocation results in fusion of the platelet-derived growth







factor receptor beta gene (PDGFRB) on chromosome 5 with this gene in a







myeloproliferative neoplasm featuring severe thrombocythemia. Alternative







splicing of this gene results in multiple transcript variants. A related pseuodgene







has been identified on chromosome 20. [provided by RefSeq, March 2012].







Transcript Variant: This variant (1) represents the shortest transcript but encodes







the longer isoform (a, also known as Kank-L). Variants 1, 3 and 4 all encode







isoform a. Sequence Note: This RefSeq record was created from transcript and







genomic sequence data to make the sequence consistent with the reference







genome assembly. The genomic coordinates used for the transcript record were







based on transcript alignments. Publication Note: This RefSeq record includes a







subset of the publications that are available for this gene. Please see the Gene







record to access additional publications. ##Evidence-Data-START## Transcript







exon combination :: AL833161.1, AK292989.1 [ECO:0000332] RNAseq







introns :: single sample supports all introns ERS025084, ERS025085







[ECO:0000348] ##Evidence-Data-END##



KAT6B
exonic
23522
histone
The protein encoded by this gene is a histone acetyltransferase and component
66





acetyltransferase
of the MOZ/MORF protein complex. In addition to its acetyltransferase activity,






KAT6B isoform 1
the encoded protein has transcriptional activation activity in its N-terminal end







and transcriptional repression activity in its C-terminal end. This protein is







necessary for RUNX2-dependent transcriptional activation and could be involved







in brain development. Mutations have been found in patients with genitopatellar







syndrome. A translocation of this gene and the CREBBP gene results in acute







myeloid leukemias. Three transcript variants encoding different isoforms have







been found for this gene. [provided by RefSeq, March 2012]. Transcript Variant:







This variant (1) represents the longest transcript and encodes the longest isoform







(1). Publication Note: This RefSeq record includes a subset of the publications







that are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







AF217500.1, BC150618.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025083, ERS025084 [ECO:0000348] ##Evidence-Data-







END##



KCTD7
exonic
154881
BTB/POZ
This gene encodes a member of the potassium channel tetramerization domain-
67





domain-
containing protein family. Family members are identified on a structural basis






containing
and contain an amino-terminal domain similar to the T1 domain present in the






protein
voltage-gated potassium channel. Mutations in this gene have been associated






KCTD7
with progressive myoclonic epilepsy-3. Alternative splicing results in multiple






isoform 1
transcript variants.[provided by RefSeq, January 2011]. Transcript Variant: This







variant (1) represents the longer transcript and encodes the longer isoform (1).







Sequence Note: This RefSeq record was created from transcript and genomic







sequence data to make the sequence consistent with the reference genome







assembly. The genomic coordinates used for the transcript record were based on







transcript alignments. ##Evidence-Data-START## Transcript exon combination ::







AK056631.1, BU902852.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025083 [ECO:0000348] ##Evidence-Data-







END##



KIF19
exonic
124602
kinesin-like
N/A
68





protein KIF19




LARP4B
exonic
23185
la-related
This gene encodes a member of an evolutionarily conserved protein family
69





protein 4B
implicated in RNA metabolism and translation. Members of this family are







characterized by the presence of an La motif, which is often located adjacent to







one or more RNA recognition motifs (RRM). Together, the two motifs constitute







the functional region of the protein and enable its interaction with the RNA







substrate. This protein family is divided into five sub-families: the genuine La







proteins and four La-related protein (LARP) sub-families. The protein encoded







by this gene belongs to LARP sub-family 4. It is a cytoplasmic protein that may







play a stimulatory role in translation. [provided by RefSeq, October 2012].







Sequence Note: This RefSeq record was created from transcript and genomic







sequence data to make the sequence consistent with the reference genome







assembly. The genomic coordinates used for the transcript record were based on







transcript alignments. ##Evidence-Data-START## CDS exon combination ::







BC152443.1, D86971.2 [ECO:0000331] RNAseq introns :: mixed/partial sample







support ERS025088 [ECO:0000350] ##Evidence-Data-END##



LOC643339
exonic
643339
N/A
N/A
70


LOH12CR1
exonic
118426
loss of
N/A
71





heterozygosity







12 chromosomal







region 1 protein




MALL
exonic
7851
MAL-like protein
This gene encodes an element of the machinery for raft-mediated trafficking in
72






endothelial cells. The encoded protein, a member of the MAL proteolipid family,







predominantly localizes in glycolipid- and cholesterol-enriched membrane







(GEM) rafts. It interacts with caveolin-1. [provided by RefSeq, July 2008].







##Evidence-Data-START## Transcript exon combination :: AK125647.1,







AK056616.1 [ECO:0000332] RNAseq introns :: single sample supports all







introns ERS025084, ERS025088 [ECO:0000348] ##Evidence-Data-END##



MAPK9
exonic
5601
mitogen-
The protein encoded by this gene is a member of the MAP kinase family. MAP
73





activated
kinases act as an integration point for multiple biochemical signals, and are






protein
involved in a wide variety of cellular processes such as proliferation,






kinase 9
differentiation, transcription regulation and development. This kinase targets






isoform alpha1
specific transcription factors, and thus mediates immediate-early gene expression







in response to various cell stimuli. It is most closely related to MAPK8, both of







which are involved in UV radiation induced apoptosis, thought to be related to







the cytochrome c-mediated cell death pathway. This gene and MAPK8 are also







known as c-Jun N-terminal kinases This kinase blocks the ubiquitination of







tumor suppressor p53, and thus it increases the stability of p53 in nonstressed







cells. Studies of this gene's mouse counterpart suggest a key role in T-cell







differentiation. Several alternatively spliced transcript variants encoding distinct







isoforms have been reported. [provided by RefSeq, September 2008]. Transcript







Variant: This variant (JNK2-a1) uses a different acceptor splice site in the last







coding exon compared to transcript variant JNK2-a2, resulting in a frameshift







and a shorter isoform (JNK2 alpha1) with a different C-terminus, compared to







isoform JNK2 alpha2. The JNK2-a1 variant differs from the JNK2-b1 variant in







the use of an alternate internal coding exon of the same length Thus, JNK2







alpha1 isoform is the same length as JNK2 beta1 isoform, with a few aa







differences in an internal protein segment. Sequence Note: This RefSeq record







was created from transcript and genomic sequence data because no single







transcript was available for the full length of the gene. The extent of this







transcript is supported by transcript alignments. Publication Note: This RefSeq







record includes a subset of the publications that are available for this gene. Please







see the Gene record to access additional publications. ##Evidence-Data-







START## CDS exon combination :: U34821.1 [ECO:0000331] RNAseq







introns :: mixed/partial sample support ERS025081, ERS025082







[ECO:0000350] ##Evidence-Data-END##



MCEE
both
84693
methylmalonyl-
The product of this gene catalyzes the interconversion of D- and L-
74





CoA epimerase,
methylmalonyl-CoA during the degradation of branched chain amino acids, odd






mitochondrial
chain-length fatty acids, and other metabolites. Mutations in this gene result in






precursor
methylmalonyl-CoA epimerase deficiency, which is presented as mild to







moderate methylmalonic aciduria. [provided by RefSeq, July 2008]. ##Evidence-







Data-START## Transcript exon combination :: BC020825.1, BG567074.1







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-END##



MGAT5
intronic
4249
alpha-1,6-
The protein encoded by this gene belongs to the glycosyltransferase family. It
75





mannosylglyco-
catalyzes the addition of beta-1,6-N-acetylglucosamine to the alpha-linked






protein 6-beta-N-
mannose of biantennary N-linked oligosaccharides present on the newly






acetyl-
synthesized glycoproteins. It is one of the most important enzymes involved in






glucosaminyl-
the regulation of the biosynthesis of glycoprotein oligosaccharides. Alterations of






transferase A
the oligosaccharides on cell surface glycoproteins cause significant changes in







the adhesive or migratory behavior of a cell. Increase in the activity of this







enzyme has been correlated with the progression of invasive malignancies.







[provided by RefSeq, October 2011]. Sequence Note: This RefSeq record was







created from transcript and genomic sequence data to make the sequence







consistent with the reference genome assembly. The genomic coordinates used







for the transcriptrecord were based on transcript alignments. Publication Note:







This RefSeq record includes a subset of the publications that are available for







this gene. Please see the Gene record to access additional publications.







##Evidence-Data-START## Transcript exon combination :: D17716.1,







AF113921.1 [ECO:0000332] RNAseq introns :: single sample supports all







introns ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-END##



MGC16275
exonic
85001
N/A
N/A
76


MGME1
exonic
92667
mitochondrial
N/A
77





genome







maintenance







exonuclease 1




MIR200A
exonic
406983

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
78






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through impeifect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications.



MIR200B
exonic
406984

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
79






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through impeifect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications.



MIR429
exonic
554210

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
80






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through impeifect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications.



MIR595
exonic
693180

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
81






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through imperfect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage.



MIR651
exonic
723779

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
82






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through impeifect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage.



MIR3163
exonic
100423029

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
83






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through impeifect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage.



MIR3910-1
exonic
100500821

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
84






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through impeifect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage.



MIR3910-2
exonic
100500902

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
85






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through imperfect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage.



MIR4267
exonic
100422994

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
86






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through imperfect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage.



MIR4436B1
exonic
100616123

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
87






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through impeifect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage.



MIR4436B2
exonic
100847033

microRNAs (miRNAs) are short (20-24 nt) non-coding RNAs that are involved
88






in post-transcriptional regulation of gene expression in multicellular organisms







by affecting both the stability and translation of mRNAs. miRNAs are







transcribed by RNA polymerase II as part of capped and polyadenylated primary







transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The







primary transcript is cleaved by the Drosha ribonuclease III enzyme to produce







an approximately 70-nt stem-loop precursor miRNA (pre-miRNA), which is







further cleaved by the cytoplasmic Dicer ribonuclease to generate the mature







miRNA and antisense miRNA star (miRNA*) products. The mature miRNA is







incorporated into a RNA-induced silencing complex (RISC), which recognizes







target mRNAs through imperfect base pairing with the miRNA and most







commonly results in translational inhibition or destabilization of the target







mRNA. The RefSeq represents the predicted microRNA stem-loop. [provided by







RefSeq, September 2009]. Sequence Note: This record represents a predicted







microRNA stem-loop as defined by miRBase. Some sequence at the 5′ and 3′







ends may not be included in the intermediate precursor miRNA produced by







Drosha cleavage.



MKL1
intronic
57591
MKL/myocardin-
The protein encoded by this gene interacts with the transcription factor
89





like protein1
myocardin, a key regulator of smooth muscle cell differentiation. The encoded







protein is predominantly nuclear and may help transduce signals from the







cytoskeleton to the nucleus. This gene is involved in a specific translocation







event that creates a fusion of this gene and the RNA-binding motif protein-15







gene. This translocation has been associated with acute megakaryocytic







leukemia. [provided by RefSeq, July 2008]. Publication Note: This RefSeq record







includes a subset of the publications that are available for this gene. Please see







the Gene record to access additional publications. ##Evidence-Data-START##







Transcript exon combination :: AB037859.2, AJ297258.1 [ECO:0000332]







RNAseq introns :: mixed/partial sample support ERS025081, ERS025082







[ECO:0000350] ##Evidence-Data-END##



MRPL42
exonic
28977
39S ribosomal
Mammalian mitochondrial ribosomal proteins are encoded by nuclear genes and
90





protein L42,
help in protein synthesis within the mitochondrion. Mitochondrial ribosomes






mitochondrial
(mitoribosomes) consist of a small 28S subunit and a large 39S subunit. They






precursor
have an estimated 75% protein to rRNA composition compared to prokaryotic







ribosomes, where this ratio is reversed. Another difference between mammalian







mitoribosomes and prokaryotic ribosomes is that the latter contain a 5S rRNA.







Among different species, the proteins comprising the mitoribosome differ greatly







in sequence, and sometimes in biochemical properties, which prevents easy







recognition by sequence homology. This gene encodes a protein identified as







belonging to both the 28S and the 39S subunits. Alternative splicing results in







multiple transcript variants. Pseudogenes corresponding to this gene are found on







chromosomes 4q, 6p, 6q, 7p, and 15q. [provided by RefSeq, May 2011].







Transcript Variant: This variant (1) encodes the supported protein. Both variants







1 and 2 encode the same protein. Sequence Note: This RefSeq record was created







from transcript and genomic sequence data to make the sequence consistent with







the reference genome assembly. The genomic coordinates used for the transcript







record were based on transcript alignments. ##RefSeq-Attributes-START## gene







product(s) localized to mito. :: reported by MitoCarta ##RefSeq-Attributes-







END## ##Evidence-Data-START## Transcript exon combination ::







AK000285.1, AF151038.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-







END##



MTHFD1
exonic
4522
C-1-
This gene encodes a protein that possesses three distinct enzymatic activities,
91





tetrahydrofolate
5,10-methylenetetrahydrofolate dehydrogenase, 5,10-methenyltetrahydrofolate






synthase,
cyclohydrolase and 10-formyltetrahydrofolate synthetase. Each of these activities






cytoplasmic
catalyzes one of three sequential reactions in the interconversion of 1-carbon







derivatives of tetrahydrofolate, which are substrates for methionine, thymidylate,







and de novo purine syntheses. The trifunctional enzymatic activities are







conferred by two major domains, an aminoterminal portion containing the







dehydrogenase and cyclohydrolase activities and a larger synthetase domain.







[provided by RefSeq, July 2008]. Publication Note: This RefSeq record includes a







subset of the publications that are available for this gene. Please see the Gene







record to access additional publications. ##RefSeq-Attributes-START## CDS







uses downstream in-frame AUG :: experimental evidence (PMID:3053686)







##RefSeq-Attributes-END## ##Evidence-Data-START## Transcript exon







combination :: BC050420.1, J04031.1 [ECO:0000332] RNAseq introns :: single







sample supports all introns ERS025081, ERS025082 [ECO:0000348]







##Evidence-Data-END##



NFIL3
exonic
4783
nuclear factor
Expression of interleukin-3 (IL3; MIM 147740) is restricted to activated T cells,
92





interleukin-3-
natural killer (NK) cells, and mast cell lines. Transcription initiation depends on






regulated
the activating capacity of specific protein factors, such as NFIL3, that bind to






protein
regulatory regions of the gene, usually upstream of the transcription start site







(Zhang et al., 1995 [PubMed 7565758]). [supplied by OMIM, February 2009].







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







S79880.1, U26173.1 [ECO:0000332] RNAseq introns :: single sample supports







all introns ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-END##



NLRP12
exonic
91662
NACHT, LRR
This gene encodes a member of the CATERPILLER family of cytoplasmic
93





and PYD
proteins. The encoded protein, which contains an N-terminal pyrin domain, a






domains-
NACHT domain, a NACHT-associated domain, and a C-terminus leucine-rich






containing
repeat region, functions as an attenuating factor of inflammation by suppressing






protein 12
inflammatory responses in activated monocytes. Mutations in this gene cause






isoform 2
familial cold autoinflammatory syndrome type 2. Alternative splicing results in







multiple transcript variants. [provided by RefSeq, March 2013]. Transcript







Variant: This variant (2) uses an alternate splice site in the central coding







region, compared to variant 3, resulting in an isoform (2) that is 1 aa shorter







than isoform 3. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to







access additional publications. ##Evidence-Data-START## Transcript







exon combination :: AY095146.1, BC028069.1 [ECO:0000332] RNAseq







introns :: single sample supports all introns ERS025089 [ECO:0000348]







##Evidence-Data-END##



NQO2
exonic
4835
ribosyldihy-
NQO2 (EC 1.10.99.2) is a flavoprotein that catalyzes the 2-electron reduction of
94





dronicotinamide
various quinones, redox dyes, and the vitamin K menadione. NQO2






dehydrogenase
predominantly uses dihydronicotinamide riboside (NRH) as the electron donor






[quinone]
(summary by Wu et al., 1997 [PubMed 9367528]). [supplied by OMIM, July







2010]. Publication Note: This RefSeq record includes a subset of the publications







that are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







J02888.1, AK311746.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-







END##



NRIP1
exonic
8204
nuclear receptor-
Nuclear receptor interacting protein 1 (NRIP1) is a nuclear protein that
95





interacting
specifically interacts with the hormone-dependent activation domain AF2 of






protein 1
nuclear receptors. Also known as RIP140, this protein modulates transcriptional







activity of the estrogen receptor. [provided by RefSeq, July 2008]. Sequence







Note: The RefSeq transcript and protein were derived from transcript and genomic







sequence to make the sequence consistent with the reference genome assembly.







The extent of this RefSeq transcript is supported by transcript alignments.







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







AK289786.1, DA230125.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025098 [ECO:0000348] ##Evidence-Data-END##



NUDT4
exonic
11163
diphosphoinositol
The protein encoded by this gene regulates the turnover of diphosphoinositol
96





polyphosphate
polyphosphates. The turnover of these high-energy diphosphoinositol






phosphohydrolase
polyphosphates represents a molecular switching activity with important






2 isoform alpha
regulatory consequences. Molecular switching by diphosphoinositol







polyphosphates may contribute to regulating intracellular trafficking. Several







alternatively spliced transcript variants have been described, but the full-length







nature of some variants has not been determined. Isoforms DIPP2alpha and







DIPP2beta are distinguishable from each other solely by DIPP2beta possessing







one additional amino acid due to intron boundary skidding in alternate splicing.







[provided by RefSeq, July 2008]. Transcript Variant: This variant (1) encodes the







predominant isoform (alpha). ##Evidence-Data-START## Transcript exon







combination :: AF191651.1, AF191650.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025081, ERS025082 [ECO:0000348]







##Evidence-Data-END##



NUDT4P1
exonic
440672
N/A
N/A
97


OVOL2
exonic
58495
transcription
N/A
98





factor







Ovo-like 2




PDE3B
intronic
5140
cGMP-inhibited
N/A
99





3′,5′-cyclic







phosphodiesterase







B




PDGFRA
exonic
5156
platelet-derived
This gene encodes a cell surface tyrosine kinase receptor for members of the
100





growth factor
platelet-derived growth factor family. These growth factors are mitogens for cells






receptor alpha
of mesenchymal origin. The identity of the growth factor bound to a receptor






precursor
monomer determines whether the functional receptor is a homodimer or a







heterodimer, composed of both platelet-derived growth factor receptor alpha and







beta polypeptides. Studies suggest that this gene plays a role in organ







development, wound healing, and tumor progression. Mutations in this gene have







been associated with idiopathic hypereosinophilic syndrome, somatic and







familial gastrointestinal stromal tumors, and a variety of other cancers. [provided







by RefSeq, March 2012]. Sequence Note: This RefSeq record was created from







transcript and genomic sequence data because no single transcript was available







for the full length of the gene. The extent of this transcript is supported by







transcript alignments and orthologous data. Publication Note: This RefSeq record







includes a subset of the publications that are available for this gene. Please see







the Gene record to access additional publications. ##Evidence-Data-START##







Transcript exon combination :: M21574.1, M22734.1 [ECO:0000332] RNAseq







introns :: single sample supports all introns ERS025081, ERS025084







[ECO:0000348] ##Evidence-Data-END##



PDSS2
exonic
57107
decaprenyl-
The protein encoded by this gene is an enzyme that synthesizes the prenyl side-
101





diphosphate
chain of coenzyme Q, or ubiquinone, one of the key elements in the respiratory






synthase
chain. The gene product catalyzes the formation of all trans-polyprenyl






subunit 2
pyrophosphates from isopentyl diphosphate in the assembly of polyisoprenoid







side chains, the first step in coenzyme Q biosynthesis. Defects in this gene are a







cause of coenzyme Q10 deficiency. [provided by RefSeq, October 2009].







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







BC039906.1, AF254956.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025084, ERS025088 [ECO:0000348] ##Evidence-







Data-END##



PHACTR4
exonic
65979
phosphatase and
This gene encodes a member of the phosphatase and actin regulator (PHACTR)
102





actin regulator
family. Other PHACTR family members have been shown to inhibit protein






4 isoform 1
phosphatase 1 (PP1) activity, and the homolog of this gene in the mouse has been







shown to interact with actin and PP1. Multiple transcript variants encoding







different isoforms have been found for this gene. [provided by RefSeq, July







2008]. Transcript Variant: This variant (1) represents the longer transcript







but encodes the shorter isoform (1). Sequence Note: This RefSeq record was







created from transcript and genomic sequence data to make the sequence







consistent with the reference genome assembly. The genomic coordinates used







for the transcript record were based on transcript alignments. ##Evidence-







Data-START## Transcript exon combination :: CR749449.1, BC029266.1







[ECO:0000332] RNAseq introns :: single sample supports all ERS025087







introns ERS025084, [ECO:0000348] ##Evidence-Data-END##



PIAS2
exonic
9063
E3 SUMO-
This gene encodes a member of the protein inhibitor of activated STAT (PIAS)
103





protein
family. PIAS proteins function as SUMO E3 ligases and play important roles in






ligase PIAS2
many cellular processes by mediating the sumoylation of target proteins.






isoform alpha
Alternatively spliced transcript variants encoding multiple isoforms have been







observed for this gene. Isoforms of the encoded protein enhance the sumoylation







of specific target proteins including the p53 tumor suppressor protein, c-Jun, and







the androgen receptor. A pseudogene of this gene is located on the short arm of







chromosome 4. The symbol MIZ1 has also been associated with ZBTB17 which







is a different gene located on chromosome 1. [provided by RefSeq, August 2011].







Transcript Variant: This variant (alpha) utilizes an alternate 3′ coding exon,







compared to variant beta, resulting in a shorter isoform (alpha) that has a unique







C-terminus compared to isoform beta. Sequence Note: This RefSeq record was







created from transcript and genomic sequence data to make the sequence







consistent with the reference genome assembly. The genomic coordinates used







for the transcript record were based on transcript alignments. Publication Note:







This RefSeq record includes a subset of the publications that are available for







this gene. Please see the Gene record to access additional publications.







##Evidence-Data-START## Transcript exon combination :: BC015190.1







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025088 [ECO:0000348] ##Evidence-Data-END##



PIK3CD
exonic
5293
phosphatidyl-
Phosphoinositide 3-kinases (PI3Ks) phosphorylate inositol lipids and are
104





inositol 4,5-
involved in the immune response. The protein encoded by this gene is a class I






bisphosphate
PI3K found primarily in leukocytes. Like other class I PI3Ks (p110-alpha p110-






3-kinase
beta, and p110-gamma), the encoded protein binds p85 adapter proteins and






catalytic subunit
GTP-bound RAS. However, unlike the other class I PI3Ks, this protein






delta isoform
phosphorylates itself, not p85 protein.[provided by RefSeq, July 2010].







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







U86453.1, Y10055.2 [ECO:0000332] RNAseq introns :: single sample supports







all introns ERS025089 [ECO:0000348] ##Evidence-Data-END##



PKHD1
intronic
5314
fibrocystin
The protein encoded by this gene is predicted to have a single transmembrane
105





isoform 1
(TM)-spanning domain and multiple copies of an immunoglobulin-like plexin-






precursor
transcription-factor domain. Alternative splicing results in two transcript variants







encoding different isoforms. Other alternatively spliced transcripts have been







described, but the full length sequences have not been determined. Several of







these transcripts are predicted to encode truncated products which lack the TM







and may be secreted. Mutations in this gene cause autosomal recessive polycystic







kidney disease, also known as polycystic kidney and hepatic disease-1. [provided







by RefSeq, July 2008]. Transcript Variant: This variant (1) encodes the longer







isoform of this protein. Publication Note: This RefSeq record includes a subset of







the publications that are available for this gene. Please see the Gene record to







access additional publications. ##Evidence-Data-START## Transcript exon







combination :: AY074797.1, AF480064.1 [ECO:0000332] RNAseq introns ::







mixed/partial sample support ERS025084, ERS025085 [ECO:0000350]







##Evidence-Data-END##



PLXNC1
exonic
10154
Plexin C1
This gene encodes a member of the plexin family. Plexins are transmembrane
106






receptors for semaphorins, a large family of proteins that regulate axon guidance,







cell motility and migration, and the immune response. The encoded protein and







its ligand regulate melanocyte adhesion, and viral semaphorins may modulate the







immune response by binding to this receptor. The encoded protein may be a







tumor suppressor protein for melanoma. Alternatively spliced transcript variants







have been observed for this gene. [provided by RefSeq, January 2011]. Transcript







Variant: This variant (2) lacks multiple 5′ exons but contains an alternate 5′ exon,







compared to variant 1. This variant is represented as non-coding due to the







presence of an upstream ORF that is predicted to interfere with translation of the







longest in-frame ORF. Translation of the upstream ORF renders the transcript a







candidate for nonsense-mediated mRNA decay (NMD). Publication Note: This







RefSeq record includes a subset of the publications that are available for this







gene. Please see the Gene record to access additional publications. ##Evidence-







Data-START## RNAseq introns :: single sample supports all introns







ERS025084, ERS025088 [ECO:0000348] ##Evidence-Data-END##



PNPLA4
exonic
8228
patatin-like
This gene encodes a member of the patatin-like family of phospholipases. The
107





phospholipase
encoded enzyme has both triacylglycerol lipase and transacylase activities and






domain-
may be involved in adipocyte triglyceride homeostasis. Alternate splicing results






containing
in multiple transcript variants. A pseudogene of this gene is found on






protein 4
chromosome Y. [provided by RefSeq, February 2010]. Transcript Variant: This






isoform 1
variant (1) represents the longest transcript and encodes the longer isoform (1).






precursor
Variants 1 and 2 encode the same isoform (1). Sequence Note: The RefSeq







transcript and protein were derived from transcript and genomic sequence to







make the sequence consistent with the reference genome assembly. The extent of







this transcript is supported by transcript alignments. ##Evidence-Data-START##







Transcript exon combination :: U03886.1, AK289888.1 [ECO:0000332] RNAseq







introns :: single sample supports all introns ERS025091, ERS025098







[ECO:0000348] ##Evidence-Data-END##



PNPT1
both
87178
polyribo-
The protein encoded by this gene belongs to the evolutionary conserved
108





nucleotide
polynucleotide phosphorylase family comprised of phosphate dependent 3′-to-5′






nucleotidyl-
exoribonucleases implicated in RNA processing and degradation. This enzyme is






transferase 1,
predominantly localized in the mitochondrial intermembrane space and is






mitochondrial
involved in import of RNA to mitochondria. Mutations in this gene have been






precursor
associated with combined oxidative phosphorylation deficiency-13 and







autosomal recessive nonsyndromic deafness-70. Related pseudogenes are found







on chromosomes 3 and 7. [provided by RefSeq, December 2012]. Publication







Note: This RefSeq record includes a subset of the publications that are







available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







BC053660.1, AJ458465.1 [ECO:0000332] RNAseq introns :: mixed/partial







sample support ERS025081, ERS025082 [ECO:0000350] ##Evidence-Data-







END## ##RefSeq-Attributes-START## gene product(s) localized to







mito. :: PMID: 12798676; reported by MitoCarta ##RefSeq-Attributes-END##



PPP2R3B
intronic
28227
serine/threonine-
Protein phosphatase 2 (formerly named type 2A) is one of the four major
109





protein
Ser/Thr phosphatases and is implicated in the negative control of cell growth and






phosphatase 2A
division. Protein phosphatase 2 holoenzymes are heterotrimeric proteins






regulatory
composed of a structural subunit A, a catalytic subunit C, and a regulatory






subunit B″
subunit B. The regulatory subunit is encoded by a diverse set of genes that have






subunit beta
been grouped into the B/PR55, B′/PR61, and B″/PR72 families. These different







regulatory subunits confer distinct enzymatic specificities and intracellular







localizations to the holozenzyme. The product of this gene belongs to the B″







family. The B″ family has been further divided into subfamilies. The product of







this gene belongs to the beta subfamily of regulatory subunit B″. [provided by







RefSeq, April 2010]. Sequence Note: This RefSeq record was created from







transcript and genomic sequence data to make the sequence consistent with the







reference genome assembly. The genomic coordinates used for the transcript







record were based on transcript alignments. ##Evidence-Data-START##







Transcript exon combination :: BK000521.1, BC063429.1 [ECO:0000332]







RNAseq introns :: single sample supports all introns ERS025084







[ECO:0000348] ##Evidence-Data-END##



PRKCB
both
5579
protein kinase C
Protein kinase C (PKC) is a family of serine- and threonine-specific protein
110





beta type
kinases that can be activated by calcium and second messenger diacylglycerol.






isoform 1
PKC family members phosphorylate a wide variety of protein targets and are







known to be involved in diverse cellular signaling pathways. PKC family







members also serve as major receptors for phorbol esters, a class of tumor







promoters. Each member of the PKC family has a specific expression profile and







is believed to play a distinct role in cells. The protein encoded by this gene is one







of the PKC family members. This protein kinase has been reported to be







involved in many different cellular functions, such as B cell activation, apoptosis







induction, endothelial cell proliferation, and intestinal sugar absorption. Studies







in mice also suggest that this kinase may also regulate neuronal functions and







correlate fear-induced conflict behavior after stress. Alternatively spliced







transcript variants encoding distinct isoforms have been reported. [provided by







RefSeq, July 2008]. Transcript Variant: This variant (1) uses an alternate splice







junction at the 5′ end of the last exon compared to variant 2. The resulting







isoform (1) has a distinct and shorter C-terminus compared to isoform 2.







Sequence Note: This RefSeq record was created from transcript and genomic







sequence data because no single transcript was available for the full length of the







gene. The extent of this transcript is supported by transcript alignments.







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







X06318.1 [ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025082, ERS025084 [ECO:0000348] ##Evidence-Data-END##



PRKCH
intronic
5583
protein kinase
Protein kinase C (PKC) is a family of serine- and threonine-specific protein
111





C eta type
kinases that can be activated by calcium and the second messenger







diacylglycerol. PKC family members phosphorylate a wide variety of protein







targets and are known to be involved in diverse cellular signaling pathways. PKC







family members also serve as major receptors for phorbol esters, a class of tumor







promoters. Each member of the PKC family has a specific expression profile and







is believed to play a distinct role in cells. The protein encoded by this gene is one







of the PKC family members. It is a calcium-independent and phospholipids-







dependent protein kinase It is predominantly expressed in epithelial tissues and







has been shown to reside specifically in the cell nucleus. This protein kinase can







regulate keratinocyte differentiation by activating the MAP kinase MAPK13







(p38delta)-activated protein kinase cascade that targets CCAAT/enhancer-







binding protein alpha (CEBPA). It is also found to mediate the transcription







activation of the transglutaminase 1 (TGM1) gene. [provided by RefSeq, July







2008]. Publication Note: This RefSeq record includes a subset of the publications







that are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







BC037268.1, AK290183.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025083 [ECO:0000348] ##Evidence-Data-







END##



PSTPIP1
exonic
9051
proline-serine-
The protein encoded by this gene binds to the cytoplasmic tail of CD2, an
112





threonine
effector of T cell activation and adhesion, negatively affecting CD2-triggered T






phosphatase-
cell activation. The encoded protein appears to be a scaffold protein and a






interacting
regulator of the actin cytoskeleton. It has also been shown to bind ABL1,






protein 1
PTPN18, WAS, CD2AP, and PTPN12. Mutations in this gene are a cause of







PAPA syndrome. [provided by RefSeq, July 2008]. Publication Note: This







RefSeq record includes a subset of the publications that are available for this







gene. Please see the Gene record to access additional publications.







##Evidence-Data-START## Transcript exon combination :: BC008602.1,







U94778.1 [ECO:0000332] RNAseq introns :: single sample supports all







introns ERS025084, ERS025088 [ECO:0000348] ##Evidence-Data-END##



PTPN2
exonic
5771
tyrosine-protein
The protein encoded by this gene is a member of the protein tyrosine
113





phosphatase non-
phosphatase (PTP) family. Members of the PTP family share a highly conserved






receptor type 2
catalytic motif, which is essential for the catalytic activity. PTPs are known to be






isoform 1
signaling molecules that regulate a variety of cellular processes including cell







growth, differentiation, mitotic cycle, and oncogenic transformation. Epidermal







growth factor receptor and the adaptor protein Shc were reported to be substrates







of this PTP, which suggested the roles in growth factor mediated cell signaling.







Multiple alternatively spliced transcript variants encoding different isoforms







have been found. Two highly related but distinctly processed pseudogenes that







localize to chromosomes 1 and 13, respectively, have been reported. [provided







by RefSeq, May 2011]. Transcript Variant: This variant (1) encodes the longest







isoform (1). Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications. ##Evidence-Data-START## Transcript exon







combination :: M25393.1, AK292570.1 [ECO:0000332] RNAseq introns ::







mixed/partial sample support ERS025081, ERS025082 [ECO:0000350]







##Evidence-Data-END##



PTPRN2
intronic
5799
receptor-type
The protein encoded by this gene is a member of the protein tyrosine
114





tyrosine-protein
phosphatase (PTP) family. PTPs are known to be signaling molecules that






phosphatase N2
regulate a variety of cellular processes including cell growth, differentiation,






isoform 1
mitotic cycle, and oncogenic transformation. This PTP possesses an extracellular






precursor
region, a single transmembrane region, and a single intracellular catalytic







domain, and thus represents a receptor-type PTP. The catalytic domain of this







PTP is most closely related to PTPRN/IA-2beta. This PTP and PTPRN are both







found to be major autoantigens associated with insulin-dependent diabetes







mellitus. Three alternatively spliced transcript variants of this gene, which







encode distinct proteins, have been reported. [provided by RefSeq, July 2008].







Transcript Variant: This variant (1) encodes the longest isoform (1). Publication







Note: This RefSeq record includes a subset of the publications that are available







for this gene. Please see the Gene record to access additional publications.







##Evidence-Data-START## Transcript exon combination :: U66702.1,







AF007555.1 [ECO:0000332] RNAseq introns :: mixed/partial sample support







ERS025081, ERS025082 [ECO:0000350] ##Evidence-Data-END##



RAB37
exonic
326624
ras-related
Rab proteins are low molecular mass GTPases that are critical regulators of
115





protein
vesicle trafficking. For additional background information on Rab proteins, see






Rab-37
MIM 179508. [supplied by OMIM, April 2006]. Transcript Variant: This variant






isoform 2
(2) represents use of an alternate promoter, 5′ UTR, and alternate start codon, and







includes an alternate coding exon, compared to variant 3. The resulting isoform







(2) has a distinct and longer N-terminus, compared to isoform 3. ##Evidence-







Data-START## Transcript exon combination :: AK098068.1, BX332255.2







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025084, ERS025088 [ECO:0000348] ##Evidence-Data-END##



RBFOX1
intronic
54715
RNA binding
The Fox-1 family of RNA-binding proteins is evolutionarily conserved, and
116





protein fox-1
regulates tissue-specific alternative splicing in metazoa. Fox-1 recognizes a






homolog 1
(U)GCAUG stretch in regulated exons or in flanking introns. The protein binds






isoform 1
to the C-terminus of ataxin-2 and may contribute to the restricted pathology of







spinocerebellar ataxia type 2 (SCA2). Ataxin-2 is the product of the SCA2 gene







which causes familial neurodegenerative diseases. Fox-1 and ataxin-2 are both







localized in the trans-Golgi network. Several alternatively spliced transcript







variants encoding different isoforms have been found for this gene. [provided by







RefSeq, November 2011]. Transcript Variant: This variant (1), also known as







gamma, encodes the longest isoform (1). Sequence Note: This RefSeq record was







created from transcript and genomic sequence data because no single transcript







was available for the full length of the gene. The extent of this transcript is







supported by transcript alignments. Publication Note: This RefSeq record includes







a subset of the publications that are available for this gene. Please see the







Gene record to access additional publications. ##Evidence-Data-START##







Transcript exon combination :: AF229057.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025083, ERS025099 [ECO:0000348]







##Evidence-Data-END##



RCC1
exonic
1104
N/A
N/A
117


RGCC
exonic
28984
regulator
This gene is thought to regulate cell cycle progression. It is induced by p53 in
118





of cell
response to DNA damage, or by sublytic levels of complement system proteins






cycle RGCC
that result in activation of the cell cycle. The encoded protein localizes to the







cytoplasm during interphase and to centrosomes during mitosis. The protein







forms a complex with polo-like kinase 1 The protein also translocates to the







nucleus in response to treatment with complement system proteins, and can







associate with and increase the kinase activity of cell division cycle 2 protein. In







different assays and cell types, overexpression of this protein has been shown to







activate or suppress cell cycle progression. [provided by RefSeq, July 2008].







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







BC066334.1, BG037019.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025081, ERS025092 [ECO:0000348] ##Evidence-Data-







END##



RHOQ
intronic
23433
rho-related GTP-
This gene encodes a member of the Rho family of small GTPases, which cycle
119





binding protein
between inactive GDP-bound and active GTP-bound states and function as






RhoQ precursor
molecular switches in signal transduction cascades. Rho proteins promote







reorganization of the actin cytoskeleton and regulate cell shape, attachment, and







motility. The encoded protein is an important signalling protein for sarcomere







assembly and has been shown to play a significant role in the exocytosis of the







solute carrier family 2, facilitated glucose transporter member 4 and other







proteins, possibly acting as the signal that turns on the membrane fusion







machinery. Three related pseudogene have been identified on chromosomes 2







and 14. [provided by RefSeq, August 2011]. Publication Note: This RefSeq







record includes a subset of the publications that are available for this gene.







Please see the Gene record to access additional publications. ##Evidence-







Data-START## Transcript exon combination :: BX428852.2, BC013135.1







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025081, ERS025082 [ECO:0000348] ##Evidence-Data-END##



RNASE3
exonic
6037
eosinophil
N/A
120





cationic







protein







precursor




RNASE10
exonic
338879
inactive
N/A
121





ribonuclease-like







protein 10







precursor




RPL38
exonic
6169
60S ribosomal
Ribosomes, the organelles that catalyze protein synthesis, consist of a small 40S
122





protein L38
subunit and a large 60S subunit. Together these subunits are composed of 4 RNA







species and approximately 80 structurally distinct proteins. This gene encodes a







ribosomal protein that is a component of the 60S subunit. The protein belongs to







the L38E family of ribosomal proteins. It is located in the cytoplasm. Alternative







splice variants have been identified, both encoding the same protein. As is typical







for genes encoding ribosomal proteins, there are multiple processed pseudogenes







of this gene dispersed through the genome, including one located in the promoter







region of the type 1 angiotensin II receptor gene. [provided by RefSeq, July







2008]. Transcript Variant: This variant (1) is the longer and predominant







transcript. Variants 1 and 2 encode the same protein. ##Evidence-Data-







START## Transcript exon combination :: BQ276548.1, BU569438.1







[ECO:0000332] RNAseq introns :: single sample supports all introns ERS025081,







ERS025082 [ECO:0000348] ##Evidence-Data-END##



RPTOR
intronic
57521
regulatory-
This gene encodes a component of a signaling pathway that regulates cell
123





associated
growth in response to nutrient and insulin levels. The encoded protein forms a






protein
stoichiometric complex with the mTOR kinase, and also associates with






of mTOR
eukaryotic initiation factor 4E-binding protein-1 and ribosomal protein S6






isoform 1
kinase. The protein positively regulates the downstream effector ribosomal







protein S6 kinase, and negatively regulates the mTOR kinase. Multiple transcript







variants encoding different isoforms have been found for this gene. [provided by







RefSeq, September 2009]. Transcript Variant: This variant (1) represents the







longer transcript and encodes the longer isoform (1). Publication Note: This







RefSeq record includes a subset of the publications that are available for this







gene. Please see the Gene record to access additional publications. ##Evidence-







Data-START## Transcript exon combination :: AY090663.1, BC136652.1







[ECO:0000332] RNAseq introns :: single sample supports all introns







ERS025083, ERS025085 [ECO:0000348] ##Evidence-Data-END##



SERPINB4
exonic
6318
serpin B4
N/A
124


SERPINB6
exonic
5269
serpin B6
The protein encoded by this gene is a member of the serpin (serine proteinase
125





isoform a
inhibitor) superfamily, and ovalbumin(ov)-selpin subfamily. It was originally







discovered as a placental thrombin inhibitor. The mouse homolog was found to







be expressed in the hair cells of the inner ear. Mutations in this gene are







associated with nonsyndromic progressive hearing loss, suggesting that this







serpin plays an important role in the inner ear in the protection against leakage







of lysosomal content during stress, and that loss of this protection results in







cell death and sensorineural hearing loss. Alternatively spliced transcript variants







have been found for this gene. [provided by RefSeq, September 2010]. Transcript







Variant: This variant (1) represents the predominant transcript. Variants 1, 5 and







6 encode the same isoform (a). Publication Note: This RefSeq record includes a







subset of the publications that are available for this gene. Please see the Gene







record to access additional publications. ##Evidence-Data-START## Transcript







exon combination :: AK314578.1, BC098564.1 [ECO:0000332] RNAseq







introns :: mixed/partial sample support ERS025081, ERS025082







[ECO:0000350] ##Evidence-Data-END##



SLC3A2
both
6520
4F2 cell-surface
This gene is a member of the solute carrier family and encodes a cell surface,
126





antigen heavy
transmembrane protein. The protein exists as the heavy chain of a heterodimer,






chain isoform b
covalently bound through di-sulfide bonds to one of several possible light chains.







The encoded transporter plays a role in regulation of intracellular calcium levels







and transports L-type amino acids. Alternatively spliced transcript variants,







encoding different isoforms, have been characterized. [provided by RefSeq,







November 2010]. Transcript Variant: This variant (2) represents the longest







transcript and encodes the longest isoform (b). Publication Note: This RefSeq







record includes a subset of the publications that are available for this gene.







Please see the Gene record to access additional publications. ##Evidence-Data-







START## Transcript exon combination :: AK025584.1 [ECO:0000332]







RNAseq introns :: mixed/partial sample support ERS025082, ERS025084







[ECO:0000350] ##Evidence-Data-END##



SLC17A5
both
26503
sialin
This gene encodes a membrane transporter that exports free sialic acids that
127






have been cleaved off of cell surface lipids and proteins from lysosomes.







Mutations in this gene cause sialic acid storage diseases, including infantile sialic







acid storage disorder and and Salla disease, an adult form. [provided by RefSeq,







July 2008]. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications. ##Evidence-Data-START## Transcript exon







combination :: BC020961.2, AJ387747.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025081, ERS025082 [ECO:0000348]







##Evidence-Data-END##



SNHG3
exonic
8420
N/A
N/A
128


SNORD17
exonic
692086
N/A
N/A
129


SNX5
exonic
27131
sorting nexin-5
This gene encodes a member of the sorting nexin family. Members of this
130





isoform a
family contain a phox (PX) domain, which is a phosphoinositide binding domain,







and are involved in intracellular trafficking. This protein functions in endosomal







sorting, the phosphoinositide-signaling pathway, and macropinocytosis. This







gene may play a role in the tumorigenesis of papillary thyroid carcinoma.







Alternative splicing results in multiple transcript variants encoding different







isoforms. [provided by RefSeq, September 2013]. Transcript Variant: This







variant (1) differs in the 5′ UTR, compared to variant 2. Variants 1 and 2







encode the same protein (isoform a). ##Evidence-Data-START## Transcript







exon combination ::BC000100.3, AF121855.1 [ECO:0000332] RNAseq introns ::







mixed/partial sample support ERS025081, ERS025082 [ECO:0000350]







##Evidence-Data-END##



SOCS2
exonic
8835
suppressor of
This gene encodes a member of the suppressor of cytokine signaling (SOCS)
131





cytokine
family. SOCS family members are cytokine-inducible negative regulators of






signaling 2
cytokine receptor signaling via the Janus kinase/signal transducer and activation







of transcription pathway (the JAK/STAT pathway). SOCS family proteins







interact with major molecules of signaling complexes to block further signal







transduction, in part, by proteasomal depletion of receptors or signal-transducing







proteins via ubiquitination. The expression of this gene can be induced by a







subset of cytokines, including erythropoietin, GM-CSF, IL10, interferon (IFN)-







gamma and by cytokine receptors such as growth horomone receptor. The







protein encoded by this gene interacts with the cytoplasmic domain of insulin-







like growth factor-1 receptor (IGF1R) and is thought to be involved in the







regulation of IGF1R mediated cell signaling. This gene has pseudogenes on







chromosomes 20 and 22. Alternative splicing results in multiple transcript







variants. [provided by RefSeq, July 2012]. Transcript Variant: This variant (1)







differs in the 5′ UTR, compared to variant 5. Variants 1-6 encode the same







protein. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications. ##Evidence-Data-START## Transcript exon







combination :: AK313165.1, AL522912.3 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025081, ERS025082 [ECO:0000348]







##Evidence-Data-END##



SOCS2-AS1
exonic
144481
N/A
N/A
132


ST8SIA5
exonic
29906
alpha-2,8-
The protein encoded by this gene is a type II membrane protein that may be
133





sialyltransferase
present in the Golgi apparatus. The encoded protein, which is a member of






8E
glycosyltransferase family 29, may be involved in the synthesis of gangliosides







GD1c, GT1a, GQ1b, and GT3 from GD1a, GT1b, GM1b, and GD3, respectively.







[provided by RefSeq, July 2008]. ##Evidence-Data-START## Transcript exon







combination :: AK056270.1, BC108910.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025082, ERS025084 [ECO:0000348]







##Evidence-Data-END##



STIM2
intronic
57620
stromal
This gene is a member of the stromal interaction molecule (STIM) family and
134





interaction
likely arose, along with related family member STIM1, from a common ancestral






molecule 2
gene. The encoded protein functions to regulate calcium concentrations in the






isoform 1
cytosol and endoplasmic reticulum, and is involved in the activation of plasma






precursor
membrane Orai Ca(2+) entry channels. This gene initiates translation from a non-







AUG (UUG) start site. A signal peptide is cleaved from the resulting protein.







Multiple transcript variants result from alternative splicing. [provided by RefSeq,







December 2009]. Transcript Variant: This variant (1) encodes the longest







isoform (1). Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to







access additional publications. ##Evidence-Data-START## Transcript







exon combination :: BC136449.1, AK096846.1 [ECO:0000332] RNAseq







introns :: single sample supports all introns ERS025081, ERS025084







[ECO:0000348] ##Evidence-Data-END## ##RefSeq-Attributes-







START## CDS uses downstream in-frame AUG ::experimental evidence







(PMID:11463338) non-AUG initiation codon :: PMID: 11463338 ##RefSeq-







Attributes-END##



TBC1D16
intronic
125058
TBC1 domain
N/A
136





family member







16 isoform a




TEX29
exonic
121793
testis-
N/A
137





expressed







sequence 29







protein




TNFRSF10A
exonic
8797
tumor necrosis
The protein encoded by this gene is a member of the TNF-receptor superfamily.
138





factor receptor
This receptor is activated by tumor necrosis factor-related apoptosis inducing






superfamily
ligand (TNFSF10/TRAIL), and thus transduces cell death signal and induces cell






member 10A
apoptosis. Studies with FADD-deficient mice suggested that FADD, a death







domain containing adaptor protein, is required for the apoptosis mediated by this







protein. [provided by RefSeq, July 2008]. Publication Note: This RefSeq record







includes a subset of the publications that are available for this gene. Please see







the Gene record to access additional publications. ##Evidence-Data-START##







Transcript exon combination :: BC012866.1, AK291299.1 [ECO:0000332]







RNAseq introns :: single sample supports all introns ERS025081, ERS025084







[ECO:0000348] ##Evidence-Data-END##



TNFRSF13C
exonic
115650
tumor necrosis
B cell-activating factor (BAFF) enhances B-cell survival in vitro and is a
139





factor receptor
regulator of the peripheral B-cell population. Overexpression of Baff in mice






superfamily
results in mature B-cell hyperplasia and symptoms of systemic lupus






member 13C
erythematosus (SLE). Also, some SLE patients have increased levels of BAFF in







serum. Therefore, it has been proposed that abnormally high levels of BAFF may







contribute to the pathogenesis of autoimmune diseases by enhancing the survival







of autoreactive B cells. The protein encoded by this gene is a receptor for BAFF







and is a type III transmembrane protein containing a single extracellular







cysteine-rich domain. It is thought that this receptor is the principal receptor







required for BAFF-mediated mature B-cell survival. [provided by RefSeq, July







2008]. Sequence Note: The RefSeq transcript and protein were derived from







genomic sequence to make the sequence consistent with the reference genome







assembly. The genomic coordinates used for the transcript record were based on







alignments. Publication Note: This RefSeq record includes a subset of the







publications that are available for this gene. Please see the Gene record to access







additional publications. ##Evidence-Data-START## Transcript exon







combination :: AF373846.1, BC112030.1 [ECO:0000332] RNAseq introns ::







single sample supports all introns ERS025089 [ECO:0000348] ##Evidence-







Data-END##



TNFRSF18
exonic
8784
tumor necrosis
This gene encodes a member of the TNF-receptor supmfamily. The encoded
140





factor receptor
receptor has been shown to have increased expression upon T-cell activation, and






superfamily
it is thought to play a key role in dominant immunological self-tolerance






member 18
maintained by CD25(+)CD4(+) regulatory T cells. Knockout studies in mice also






isoform
suggest the role of this receptor is in the regulation of CD3-driven T-cell






1 precursor
activation and programmed cell death. Three alternatively spliced transcript







variants of this gene encoding distinct isoforms have been reported. [provided by







RefSeq, February 2011]. Transcript Variant: This variant (1) represents the







longest transcript. It contains an extra coding segment, which leads to a frame







shift, compared to variant 2. The resulting preotein (isoform 1) contains a







distinct and shorter C-terminus, as compared to isoform 2. Sequence Note: This







RefSeq record was created from transcript and genomic sequence data to make







the sequence consistent with the reference genome assembly. The genomic







coordinates used for the transcript record were based on transcript alignments.







Publication Note: This RefSeq record includes a subset of the publications that







are available for this gene. Please see the Gene record to access additional







publications. ##Evidence-Data-START## Transcript exon combination ::







AY358877.1, AF125304.1 [ECO:0000332] RNAseq introns :: single sample







supports all introns ERS025089, ERS025093 [ECO:0000348] ##Evidence-Data-







END##



TRAFD1
exonic
10906
TRAF-type zinc
The innate immune system confers host defense against viral and microbial
141





finger domain-
infection, and TRAFD1 is a negative feedback regulator that controls excessive






containing
immune responses (Sanada et al., 2008 [PubMed 18849341]). [supplied by






protein 1
OMIM, December 2009]. Transcript Variant: This variant (1) represents the







longer transcript. Variants 1 and 2 both encode the same protein. ##Evidence-







Data-START## Transcript exon combination :: AK122620.1 [ECO:0000332]







RNAseq introns :: single sample supports all introns ERS025083, ERS025084







[ECO:0000348] ##Evidence-Data-END##



TRPM2
exonic
7226
Transient
The protein encoded by this gene is a calcium-permeable cation channel that is
142





Receptor
regulated by free intracellular ADP-ribose. The encoded protein is activated by






Potential
oxidative stress and confers susceptibility to cell death Several alternatively






Cation Channel
spliced transcript variants of this gene have been described, but their full-length






Subfamily
nature is not known. [provided by RefSeq, July 2008]. Transcript Variant: This






M Member 2
variant (2) uses an alternate in-frame splice junction at the 5′ end of an exon







compared to variant 1. This results in the introduction of a premature stop codon







and renders the transcript a nonsense-mediated mRNA decay (NMD) candidate.







Therefore, this transcript is not thought to be protein-coding. Publication Note:







This RefSeq record includes a subset of the publications that are available for







this gene. Please see the Gene record to access additional publications.







##Evidence-Data-START## RNAseq introns :: mixed/partial sample support







ERS025081, ERS025082 [ECO:0000350] ##Evidence-Data-END##



TTLL10
exonic
254173
inactive
N/A
143





polyglycylase







TTLL10







isoform 1




TTYH2
exonic
94015
protein tweety
This gene encodes a member of the tweety family of proteins. Members of this
144





homolog 2
family function as chloride anion channels. The encoded protein functions as a






isoform 1
calcium(2+)-activated large conductance chloride(−) channel, and may play a role







in kidney tumorigenesis. Two transcript variants encoding distinct isoforms have







been identified for this gene. [provided by RefSeq, July 2008]. Transcript Variant:







This variant (1) represents the longer transcript, and encodes the longer isoform







(1). ##Evidence-Data-START## Transcript exon combination :: AF319952.1,







BC107492.1 [ECO:0000332] RNAseq introns :: single sample supports all







introns ERS025082, ERS025083 [ECO:0000348] ##Evidence-Data-END##



UBE2N
exonic
7334
ubiquitin-
The modification of proteins with ubiquitin is an important cellular mechanism
145





conjugating
for targeting abnormal or short-lived proteins for degradation. Ubiquitination






enzyme E2 N
involves at least three classes of enzymes: ubiquitin-activating enzymes, or E1s,







ubiquitin-conjugating enzymes, or E2s, and ubiquitin-protein ligases, or E3s.







This gene encodes a member of the E2 ubiquitin-conjugating enzyme family.







Studies in mouse suggest that this protein plays a role in DNA postreplication







repair. [provided by RefSeq, July 2008]. Publication Note: This RefSeq record







includes a subset of the publications that are available for this gene. Please see







the Gene record to access additional publications. ##Evidence-Data-START##







Transcript exon combination :: BC000396.2, D83004.1 [ECO:0000332] RNAseq







introns :: single sample supports all introns ERS025084 [ECO:0000348]







##Evidence-Data-END##



VCX
exonic
26609
variable charge
This gene belongs to the VCX/Y gene family, which has multiple members on
146





X-linked
both X and Y chromosomes, and all are expressed exclusively in male germ






protein 1
cells. The X-linked members are clustered on chromosome Xp22 and Y-linked







members are two identical copies of the gene within a palindromic region on







Yq11. The family members share a high degree of sequence identity, with the







exception that a 30-bp unit is tandemly repeated in X-linked members but occurs







only once in Y-linked members. The VCX gene cluster is polymorphic in terms







of copy number; different individuals may have a different number of VCX







genes. VCX/Y genes encode small and highly charged proteins of unknown







function. The presence of a putative bipartite nuclear localization signal suggests







that VCX/Y members are nuclear proteins. This gene contains 10 repeats of the







30-bp unit. [provided by RefSeq, July 2008] ##Evidence-Data-START##







Transcript exon combination :: AF167081.2 [ECO:0000332] ##Evidence-Data-







END##



VSTM1
intronic
284415
V-set and
N/A
147





transmembrane







domain-







containing







protein 1







precursor




VWA2
exonic
340706
von Willebrand
This gene encodes a member of the von Willebrand factor A-like domain protein
148





factor A domain-
superfamily. The encoded protein is localized to the extracellular matrix and may






containing
serve as a structural component in basement membranes or in anchoring






protein 2
structures on scaffolds of collagen VII or fibrillin. This gene has been linked to






precursor
type 1A diabetes and is a candidate serological marker for colon cancer.







[provided by RefSeq, January 2013]. Sequence Note: This RefSeq record was







created from transcript and genomic sequence data to make the sequence







consistent with the reference genome assembly. The genomic coordinates used for







the transcript record were based on transcript alignments. CCDS Note: The coding







region has been updated to represent an alternative 3′ splicing pattern that







is more supported by the available transcript and protein data ##Evidence-







Data-START## Transcript exon combination :: AY572972.1, AJ536328.2







[ECO:0000332] RNAseq introns :: mixed/partial sample support ERS025081,







ERS025084 [ECO:0000350] ##Evidence-Data-END##



ZNF350
exonic
59348
zinc finger
N/A
149





protein 350




ZNF432
exonic
9668
zinc finger
N/A
150





protein 432




ZNF577
exonic
84765
N/A
N/A
151


ZNF613
exonic
79898
zinc finger
N/A
152





protein 613







isoform 1




ZNF614
exonic
80110
zinc finger
N/A
153





protein 614




ZNF615
exonic
284370
zinc finger
N/A
154





protein 615







isoform 1




ZNF649
exonic
65251
zinc finger
N/A
155





protein 649




ZNF841
exonic
284371
zinc finger
N/A
156





protein 841









For all genes listed in Table 2 (namely, those relevant to CNV-subregions of interest), Table 3 represents a non-redundant list.









TABLE 4







A non-redundant list of transcript variants that correspond to the genes in Table 3











RefSeq

RefSeq




Gene
Exon
Accession

SEQ


Symbol
overlap
Number
mRNA_Description
ID














MIR200B
exonic
NR_029639

Homo sapiens microRNA 200b (MIR200B), microRNA.

173


MIR200A
exonic
NR 029834

Homo sapiens microRNA 200a (MIR200A), microRNA.

174


MIR429
exonic
NR 029957

Homo sapiens microRNA 429 (MIR429), microRNA.

175


TTLL10
exonic
NM 001130045

Homo sapiens tubulin tyrosine ligase-like family, member 10 (TTLL10), transcript variant 1,

176





mRNA.



TTLL10
exonic
NM_153254

Homo sapiens tubulin tyrosine ligase-like family, member 10 (TTLL10), transcript variant 2,

177





mRNA.



TNFRSF18
exonic
NM_004195

Homo sapiens tumor necrosis factor receptor superfamily, member 18 (TNFRSF18), transcript

178





variant 1, mRNA.



TNFRSF18
exonic
NM 148901

Homo sapiens tumor necrosis factor receptor superfamily, member 18 (TNFRSF18), transcript

179





variant 2, mRNA.



TNFRSF18
exonic
NM_148902

Homo sapiens tumor necrosis factor receptor superfamily, member 18 (TNFRSF18), transcript

180





variant 3, mRNA.



PIK3CD
exonic
NM_005026

Homo sapiens phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit

181





delta (PIK3CD), mRNA.



CAPZB
intronic
NR 038125

Homo sapiens capping protein (actin filament) muscle Z-line, beta (CAPZB),

182





transcript variant 4, non-coding RNA.



CAPZB
intronic
NM 001206540

Homo sapiens capping protein (actin filament) muscle Z-line, beta (CAPZB),

183





transcript variant 2, mRNA.



CAPZB
intronic
NM_004930

Homo sapiens capping protein (actin filament) muscle Z-line, beta (CAPZB),

184





transcript variant 1, mRNA.



IFNLR1
exonic
NM 170743

Homo sapiens interferon, lambda receptor 1 (IFNLR1), transcript variant 1, mRNA.

185


IFNLR1
exonic
NM_173064

Homo sapiens interferon, lambda receptor 1 (IFNLR1), transcript variant 2, mRNA.

186


IFNLR1
exonic
NM 173065

Homo sapiens interferon, lambda receptor 1 (IFNLR1), transcript variant 3, mRNA.

187


PHACTR4
exonic
NM 001048183

Homo sapiens phosphatase and actin regulator 4 (PHACTR4), transcript variant 1, mRNA.

188


PHACTR4
exonic
NM_023923

Homo sapiens phosphatase and actin regulator 4 (PHACTR4), transcript variant 2, mRNA.

189


SNHG3
exonic
NR 002909

Homo sapiens small nucleolar RNA host gene 3 (non-protein coding) (SNHG3), transcript

190





variant 2, non-coding RNA.



SNHG3
exonic
NR 036473

Homo sapiens small nucleolar RNA host gene 3 (non-protein coding) (SNHG3), transcript

191





variant 1, non-coding RNA.



RCC1
exonic
NM 001048199

Homo sapiens regulator of chromosome condensation 1 (RCC1), transcript variant 4, mRNA.

192


RCC1
exonic
NR 030725

Homo sapiens regulator of chromosome condensation 1 (RCC1), transcript variant 5,

193





non-coding RNA.



RCC1
exonic
NR_030726

Homo sapiens regulator of chromosome condensation 1 (RCC1), transcript variant 6,

194





non-coding RNA.



RCC1
exonic
NM_001048194

Homo sapiens regulator of chromosome condensation 1 (RCC1), transcript variant 1, mRNA.

195


RCC1
exonic
NM 001048195

Homo sapiens regulator of chromosome condensation 1 (RCC1), transcript variant 2, mRNA.

196


RCC1
exonic
NM 001269

Homo sapiens regulator of chromosome condensation 1 (RCC1), transcript variant 3, mRNA.

197


AGBL4
intronic
NM_032785

Homo sapiens ATP/GTP binding protein-like 4 (AGBL4), mRNA.

198


GPATCH2
intronic
NM_018040

Homo sapiens G patch domain containing 2 (GPATCH2), mRNA.

199


RHOQ
intronic
NM 012249

Homo sapiens ras homolog family member Q (RHOQ), mRNA.

200


PNPT1
both
NM 033109

Homo sapiens polyribonucleotide nucleotidyltransferase 1 (PNPT1), mRNA.

201


MCEE
both
NM 032601

Homo sapiens methylmalonyl CoA epimerase (MCEE), mRNA.

202


HK2
exonic
NM_000189

Homo sapiens hexokinase 2 (HK2), mRNA.

203


FHL2
exonic
NM_201557

Homo sapiens four and a half LIM domains 2 (FHL2), transcript variant 4, mRNA.

204


FHL2
intronic
NM 001039492

Homo sapiens four and a half LIM domains 2 (FHL2), transcript variant 5, mRNA.

205


FHL2
intronic
NM 001450

Homo sapiens four and a half LIM domains 2 (FHL2), transcript variant 1, mRNA.

206


FHL2
intronic
NM 201555

Homo sapiens four and a half LIM domains 2 (FHL2), transcript variant 2, mRNA.

207


MIR4267
exonic
NR_036225

Homo sapiens microRNA 4267 (MIR4267), microRNA.

208


MALL
exonic
NM_005434

Homo sapiens mal, T-cell differentiation protein-like (MALL), mRNA.

209


MIR4436B1
exonic
NR 039941

Homo sapiens microRNA 4436b-1 (MIR4436B1), microRNA.

210


MIR4436B2
exonic
NR 049830

Homo sapiens microRNA 4436b-2 (MIR4436B2), microRNA.

211


MGAT5
intronic
NM 002410

Homo sapiens mannosyl (alpha-1,6-)-glycoprotein beta-1,6-N-acetyl-glucosaminyltransferase

212





(MGAT5), mRNA.



BMPR2
intronic
NM_001204

Homo sapiens bone morphogenetic protein receptor, type II (serine/threonine kinase)

213





(BMPR2), mRNA.



DNER
intronic
NM 139072

Homo sapiens delta/notch-like EGF repeat containing (DNER), mRNA.

214


IQCB1
exonic
NM 001023570

Homo sapiens IQ motif containing B1 (IQCB1), transcript variant 1, mRNA.

215


IQCB1
exonic
NM 001023571

Homo sapiens IQ motif containing B1 (IQCB1), transcript variant 3, mRNA.

216


STIM2
intronic
NM_001169117

Homo sapiens stromal interaction molecule 2 (STIM2), transcript variant 3, mRNA.

217


STIM2
intronic
NM_001169118

Homo sapiens stromal interaction molecule 2 (STIM2), transcript variant 1, mRNA.

218


STIM2
intronic
NM 020860

Homo sapiens stromal interaction molecule 2 (STIM2), transcript variant 2, mRNA.

219


PDGFRA
exonic
NM 006206

Homo sapiens platelet-derived growth factor receptor, alpha polypeptide (PDGFRA), mRNA.

220


HCN1
exonic
NM_021072

Homo sapiens hyperpolarization activated cyclic nucleotide-gated potassium channel 1

221





(HCN1), mRNA.



EMB
exonic
NM 198449

Homo sapiens embigin (EMB), mRNA.

222


EDIL3
intronic
NM_005711

Homo sapiens EGF-like repeats and discoidin I-like domains 3 (EDIL3), transcript variant 1,

223





mRNA.



MAPK9
exonic
NM_002752

Homo sapiens mitogen-activated protein kinase 9 (MAPK9), transcript variant JNK2-a2,

224





mRNA.



MAPK9
exonic
NM 139068

Homo sapiens mitogen-activated protein kinase 9 (MAPK9), transcript variant JNK2-a1,

225





mRNA.



MAPK9
exonic
NM 139069

Homo sapiens mitogen-activated protein kinase 9 (MAPK9), transcript variant JNK2-b1,

226





mRNA.



MAPK9
exonic
NM_139070

Homo sapiens mitogen-activated protein kinase 9 (MAPK9), transcript variant JNK2-b2,

227





mRNA.



MAPK9
exonic
NM_001135044

Homo sapiens mitogen-activated protein kinase 9 (MAPK9), transcript variant JNK2-g,

228





mRNA.



SERPINB6
exonic
NM 001271825

Homo sapiens serpin peptidase inhibitor, clade B (ovalbumin), member 6 (SERPINB6),

229





transcript variant 6, mRNA.



SERPINB6
exonic
NM 001271823

Homo sapiens serpin peptidase inhibitor, clade B (ovalbumin), member 6 (SERPINB6),

230





transcript variant 4, mRNA.



SERPINB6
exonic
NM_001271822

Homo sapiens serpin peptidase inhibitor, clade B (ovalbumin), member 6 (SERPINB6),

231





transcript variant 3, mRNA.



SERPINB6
exonic
NM_001195291

Homo sapiens serpin peptidase inhibitor, clade B (ovalbumin), member 6 (SERPINB6),

232





transcript variant 2, mRNA.



SERPINB6
exonic
NM 001271824

Homo sapiens serpin peptidase inhibitor, clade B (ovalbumin), member 6 (SERPINB6),

233





transcript variant 5, mRNA.



SERPINB6
exonic
NM 004568

Homo sapiens serpin peptidase inhibitor, clade B (ovalbumin), member 6 (SERPINB6),

234





transcript variant 1, mRNA.



DKFZP686I15217
exonic
NR_026855

Homo sapiens long intergenic non-protein coding RNA 1011 (LINC01011), transcript

235





variant 1, non-coding RNA.



DKFZP686I15217
exonic
NR_026856

Homo sapiens long intergenic non-protein coding RNA 1011 (LINC01011), transcript

236





variant 2, non-coding RNA.



NQO2
exonic
NM_000904

Homo sapiens NAD(P)H dehydrogenase, quinone 2 (NQO2), mRNA.

237


HTATSF1P2
exonic
NR 033884

Homo sapiens HIV-1 Tat specific factor 1 pseudogene 2 (HTATSF1P2), non-coding RNA.

238


PKHD1
intronic
NM_138694

Homo sapiens polycystic kidney and hepatic disease 1 (autosomal recessive) (PKHD1),

239





transcript variant 1, mRNA.



PKHD1
intronic
NM_170724

Homo sapiens polycystic kidney and hepatic disease 1 (autosomal recessive) (PKHD1),

240





transcript variant 2, mRNA.



SLC17A5
both
NM 012434

Homo sapiens solute carrier family 17 (acidic sugar transporter), member 5 (SLC17A5),

241





mRNA.



PDSS2
exonic
NM_020381

Homo sapiens prenyl (decaprenyl) diphosphate synthase, subunit 2 (PDSS2), mRNA.

242


KCTD7
exonic
NM 001167961

Homo sapiens potassium channel tetramerization domain containing 7 (KCTD7), transcript

243





variant 2, mRNA.



KCTD7
exonic
NM 153033

Homo sapiens potassium channel tetramerization domain containing 7 (KCTD7), transcript

244





variant 1, mRNA.



PTPRN2
intronic
NM_002847

Homo sapiens protein tyrosine phosphatase, receptor type, N polypeptide 2 (PTPRN2),

245





transcript variant 1, mRNA.



PTPRN2
intronic
NM_130842

Homo sapiens protein tyrosine phosphatase, receptor type, N polypeptide 2 (PTPRN2),

246





transcript variant 2, mRNA.



PTPRN2
intronic
NM 130843

Homo sapiens protein tyrosine phosphatase, receptor type, N polypeptide 2 (PTPRN2),

247





transcript variant 3, mRNA.



MIR595
exonic
NR 030325

Homo sapiens microRNA 595 (MIR595), microRNA.

248


TNFRSF10A
exonic
NM 003844

Homo sapiens tumor necrosis factor receptor superfamily, member 10a (TNFRSF10A),

249





mRNA.



IDO2
intronic
NM 194294

Homo sapiens indoleamine 2,3-dioxygenase 2 (IDO2), mRNA.

250


STK3
intronic
NM_001256313

Homo sapiens serine/threonine kinase 3 (STK3), transcript variant 3, mRNA.

251


STK3
intronic
NM_006281

Homo sapiens serine/threonine kinase 3 (STK3), transcript variant 1, mRNA.

252


STK3
intronic
NM 001256312

Homo sapiens serine/threonine kinase 3 (STK3), transcript variant 2, mRNA.

253


KANK1
intronic
NM_001256876

Homo sapiens KN motif and ankyrin repeat domains 1 (KANK1), transcript variant 3, mRNA.

254


KANK1
intronic
NM_001256877

Homo sapiens KN motif and ankyrin repeat domains 1 (KANK1), transcript variant 4, mRNA.

255


KANK1
intronic
NM 015158

Homo sapiens KN motif and ankyrin repeat domains 1 (KANK1), transcript variant 1, mRNA.

256


KANK1
intronic
NM 153186

Homo sapiens KN motif and ankyrin repeat domains 1 (KANK1), transcript variant 2, mRNA.

257


GDA
exonic
NM 001242507

Homo sapiens guanine deaminase (GDA), transcript variant 4, mRNA.

258


GDA
exonic
NM 001242505

Homo sapiens guanine deaminase (GDA), transcript variant 1, mRNA.

259


GDA
exonic
NM_001242506

Homo sapiens guanine deaminase (GDA), transcript variant 3, mRNA.

260


GDA
exonic
NM_004293

Homo sapiens guanine deaminase (GDA), transcript variant 2, mRNA.

261


AUH
exonic
NM 001698

Homo sapiens AU RNA binding protein/enoyl-CoA hydratase (AUH), mRNA.

262


MIR3163
exonic
NR 036121

Homo sapiens microRNA 3163 (MIR3163), microRNA.

263


NFIL3
exonic
NM 005384

Homo sapiens nuclear factor, interleukin 3 regulated (NFIL3), mRNA.

264


MIR3910-1
exonic
NR_037472

Homo sapiens microRNA 3910-1 (MIR3910-1), microRNA.

265


MIR3910-2
exonic
NR_037489

Homo sapiens microRNA 3910-2 (MIR3910-2), microRNA.

266


ASTN2
intronic
NM 014010

Homo sapiens astrotactin 2 (ASTN2), transcript variant 1, mRNA.

267


ASTN2
intronic
NM_198186

Homo sapiens astrotactin 2 (ASTN2), transcript variant 2, mRNA.

268


ASTN2
intronic
NM 001184734

Homo sapiens astrotactin 2 (ASTN2), transcript variant 5, mRNA.

269


ASTN2
intronic
NM 198187

Homo sapiens astrotactin 2 (ASTN2), transcript variant 3, mRNA.

270


ASTN2
intronic
NM_198188

Homo sapiens astrotactin 2 (ASTN2), transcript variant 4, mRNA.

271


ASTN2
intronic
NM_001184735

Homo sapiens astrotactin 2 (ASTN2), transcript variant 6, mRNA.

272


LARP4B
exonic
NM 015155

Homo sapiens La ribonucleoprotein domain family, member 4B (LARP4B), mRNA.

273


GTPBP4
exonic
NM 012341

Homo sapiens GTP binding protein 4 (GTPBP4), mRNA.

274


IDI2
exonic
NM 033261

Homo sapiens isopentenyl-diphosphate delta isomerase 2 (IDI2), mRNA.

275


IDI2-AS1
exonic
NR_024628

Homo sapiens IDI2 antisense RNA 1 (IDI2-AS1), transcript variant 1, non-coding RNA.

276


IDI2-AS1
exonic
NR_024629

Homo sapiens IDI2 antisense RNA 1 (IDI2-AS1), transcript variant 2, non-coding RNA.

277


IDI2-AS1
exonic
NR 027708

Homo sapiens IDI2 antisense RNA 1 (IDI2-AS1), transcript variant 3, non-coding RNA.

278


IDI2-AS1
exonic
NR 027709

Homo sapiens IDI2 antisense RNA 1 (IDI2-AS1), transcript variant 4, non-coding RNA.

279


KAT6B
exonic
NM 001256468

Homo sapiens K(lysine) acetyltransferase 6B (KAT6B), transcript variant 2, mRNA.

280


KAT6B
exonic
NM_001256469

Homo sapiens K(lysine) acetyltransferase 6B (KAT6B), transcript variant 3, mRNA.

281


KAT6B
exonic
NM_012330

Homo sapiens K(lysine) acetyltransferase 6B (KAT6B), transcript variant 1, mRNA.

282


VWA2
exonic
NM 001272046

Homo sapiens von Willebrand factor A domain containing 2 (VWA2), mRNA.

283


PDE3B
intronic
NM 000922

Homo sapiens phosphodiesterase 3B, cGMP-inhibited (PDE3B), mRNA.

284


EHF
intronic
NM_001206615

Homo sapiens ets homologous factor (EHF), transcript variant 3, mRNA.

285


EHF
intronic
NM_012153

Homo sapiens ets homologous factor (EHF), transcript variant 2, mRNA.

286


EHF
exonic
NM_001206616

Homo sapiens ets homologous factor (EHF), transcript variant 1, mRNA.

287


SLC3A2
exonic
NM_001012662

Homo sapiens solute carrier family 3 (amino acid transporter heavy chain), member 2

288





(SLC3A2), transcript variant 2, mRNA.



SLC3A2
intronic
NM 001012664

Homo sapiens solute carrier family 3 (amino acid transporter heavy chain), member 2

289





(SLC3A2), transcript variant 5, mRNA.



SLC3A2
exonic
NM 002394

Homo sapiens solute carrier family 3 (amino acid transporter heavy chain), member 2

290





(SLC3A2), transcript variant 3, mRNA.



SLC3A2
intronic
NM_001013251

Homo sapiens solute carrier family 3 (amino acid transporter heavy chain), member 2

291





(SLC3A2), transcript variant 6, mRNA.



SLC3A2
intronic
NR_037193

Homo sapiens solute carrier family 3 (amino acid transporter heavy chain), member 2

292





(SLC3A2), transcript variant 7, non-coding RNA.



GDPD4
exonic
NM 182833

Homo sapiens glycerophosphodiester phosphodiesterase domain containing 4 (GDPD4),

293





mRNA.



ETV6
exonic
NM_001987

Homo sapiens ets variant 6 (ETV6), mRNA.

294


LOH12CR1
exonic
NM_058169

Homo sapiens loss of heterozygosity, 12, chromosomal region 1 (LOH12CR1), mRNA.

295


DUSP16
exonic
NM 030640

Homo sapiens dual specificity phosphatase 16 (DUSP16), mRNA.

296


CREBL2
exonic
NM 001310

Homo sapiens cAMP responsive element binding protein-like 2 (CREBL2), mRNA.

297


GPR19
exonic
NM 006143

Homo sapiens G protein-coupled receptor 19 (GPR19), mRNA.

298


CDKN1B
exonic
NM_004064

Homo sapiens cyclin-dependent kinase inhibitor 1B (p27, Kip1) (CDKN1B), mRNA.

299


APOLD1
exonic
NM_001130415

Homo sapiens apolipoprotein L domain containing 1 (APOLD1), transcript variant 1, mRNA.

300


APOLD1
intronic
NM 030817

Homo sapiens apolipoprotein L domain containing 1 (APOLD1), transcript variant 2, mRNA.

301


EEA1
exonic
NM 003566

Homo sapiens early endosome antigen 1 (EEA1), mRNA.

302


LOC643339
exonic
NR 040096

Homo sapiens uncharacterized LOC643339 (LOC643339), non-coding RNA.

303


NUDT4
exonic
NM 019094

Homo sapiens nudix (nucleoside diphosphate linked moiety X)-type motif 4 (NUDT4),

304





transcript variant 1, mRNA.



NUDT4
exonic
NM_199040

Homo sapiens nudix (nucleoside diphosphate linked moiety X)-type motif 4 (NUDT4),

305





transcript variant 2, mRNA.



NUDT4P1
exonic
NR_002212

Homo sapiens nudix (nucleoside diphosphate linked moiety X)-type motif 4 pseudogene 1

306





(NUDT4P1), non-coding RNA.



UBE2N
exonic
NM_003348

Homo sapiens ubiquitin-conjugating enzyme E2N (UBE2N), mRNA.

307


MRPL42
exonic
NM 014050

Homo sapiens mitochondrial ribosomal protein L42 (MRPL42), transcript variant 1, mRNA.

308


MRPL42
exonic
NM 172177

Homo sapiens mitochondrial ribosomal protein L42 (MRPL42), transcript variant 2, mRNA.

309


MRPL42
exonic
NR 038159

Homo sapiens mitochondrial ribosomal protein L42 (MRPL42), transcript variant 3,

310





non-coding RNA.



MRPL42
exonic
NR_038160

Homo sapiens mitochondrial ribosomal protein L42 (MRPL42), transcript variant 4,

311





non-coding RNA.



MRPL42
exonic
NR 038161

Homo sapiens mitochondrial ribosomal protein L42 (MRPL42), transcript variant 5,

312





non-coding RNA.



SOCS2-AS1
exonic
NR_038263

Homo sapiens SOCS2 antisense RNA 1 (SOCS2-AS1), non-coding RNA.

313


SOCS2
exonic
NM_003877

Homo sapiens suppressor of cytokine signaling 2 (SOCS2), transcript variant 1, mRNA.

314


SOCS2
exonic
NM 001270467

Homo sapiens suppressor of cytokine signaling 2 (SOCS2), transcript variant 2, mRNA.

315


SOCS2
exonic
NM 001270468

Homo sapiens suppressor of cytokine signaling 2 (SOCS2), transcript variant 3, mRNA.

316


SOCS2
exonic
NM_001270469

Homo sapiens suppressor of cytokine signaling 2 (SOCS2), transcript variant 4, mRNA.

317


SOCS2
exonic
NM_001270470

Homo sapiens suppressor of cytokine signaling 2 (SOCS2), transcript variant 5, mRNA.

318


SOCS2
exonic
NM_001270471

Homo sapiens suppressor of cytokine signaling 2 (SOCS2), transcript variant 6, mRNA.

319


CRADD
exonic
NM_003805

Homo sapiens CASP2 and RIPK1 domain containing adaptor with death domain (CRADD),

320





mRNA.



PLXNC1
exonic
NM_005761

Homo sapiens plexin C1 (PLXNC1), transcript variant 1, mRNA.

321


PLXNC1
exonic
NR 037687

Homo sapiens plexin C1 (PLXNC1), transcript variant 2, non-coding RNA.

322


CCDC41
exonic
NM 001042399

Homo sapiens coiled-coil domain containing 41 (CCDC41), transcript variant 2, mRNA.

323


CCDC41
exonic
NM_016122

Homo sapiens coiled-coil domain containing 41 (CCDC41), transcript variant 1, mRNA.

324


TRAFD1
exonic
NM 001143906

Homo sapiens TRAF-type zinc finger domain containing 1 (TRAFD1), transcript variant 1,

325





mRNA.



TRAFD1
exonic
NM 006700

Homo sapiens TRAF-type zinc finger domain containing 1 (TRAFD1), transcript variant 2,

326





mRNA.



RGCC
exonic
NM 014059

Homo sapiens regulator of cell cycle (RGCC), mRNA.

327


COMMD6
exonic
NM 203495

Homo sapiens COMM domain containing 6 (COMMD6), transcript variant 2, mRNA.

328


COMMD6
exonic
NM_203497

Homo sapiens COMM domain containing 6 (COMMD6), transcript variant 1, mRNA.

329


GPC5
intronic
NM_004466

Homo sapiens glypican 5 (GPC5), mRNA.

330


ARHGEF7
exonic
NM_003899

Homo sapiens Rho guanine nucleotide exchange factor (GEF) 7 (ARHGEF7), transcript

331





variant 1, mRNA.



ARHGEF7
exonic
NM 001113513

Homo sapiens Rho guanine nucleotide exchange factor (GEF) 7 (ARHGEF7), transcript

332





variant 5, mRNA.



TEX29
exonic
NM_152324

Homo sapiens testis expressed 29 (TEX29), mRNA.

333


ARHGEF7
intronic
NM 001113511

Homo sapiens Rho guanine nucleotide exchange factor (GEF) 7 (ARHGEF7), transcript

334





variant 3, mRNA.



ARHGEF7
intronic
NM 001113512

Homo sapiens Rho guanine nucleotide exchange factor (GEF) 7 (ARHGEF7), transcript

335





variant 4, mRNA.



ARHGEF7
intronic
NM_145735

Homo sapiens Rho guanine nucleotide exchange factor (GEF) 7 (ARHGEF7), transcript

336





variant 2, mRNA.



RNASE10
exonic
NM_001012975

Homo sapiens ribonuclease, RNase A family, 10 (non-active) (RNASE10), mRNA.

337


RNASE3
exonic
NM 002935

Homo sapiens ribonuclease, RNase A family, 3 (RNASE3), mRNA.

338


ECRP
exonic
NR_033909

Homo sapiens ribonuclease, RNase A family, 2 (liver, eosinophil-derived neurotoxin)

339





pseudogene (ECRP), non-coding RNA.



PRKCH
intronic
NM 006255

Homo sapiens protein kinase C, eta (PRKCH), mRNA.

340


MTHFD1
exonic
NM 005956

Homo sapiens methylenetetrahydrofolate dehydrogenase (NADP+ dependent) 1,

341





methenyltetrahydrofolate cyclohydrolase, formyltetrahydrofolate synthetase (MTHFD1),






mRNA.



BDKRB2
intronic
NM 000623

Homo sapiens bradykinin receptor B2 (BDKRB2), mRNA.

342


HEXA
exonic
NM_000520

Homo sapiens hexosaminidase A (alpha polypeptide) (HEXA), mRNA.

343


PSTPIP1
exonic
NM_003978

Homo sapiens proline-serine-threonine phosphatase interacting protein 1 (PSTPIP1), mRNA.

344


RBFOX1
intronic
NM_001142333

Homo sapiens RNA binding protein, fox-1 homolog (C. elegans) 1 (RBFOX1), transcript

345





variant 5, mRNA.



RBFOX1
intronic
NM 018723

Homo sapiens RNA binding protein, fox-1 homolog (C. elegans) 1 (RBFOX1), transcript

346





variant 4, mRNA.



RBFOX1
intronic
NM 001142334

Homo sapiens RNA binding protein, fox-1 homolog (C. elegans) 1 (RBFOX1), transcript

347





variant 6, mRNA.



RBFOX1
intronic
NM_145891

Homo sapiens RNA binding protein, fox-1 homolog (C. elegans) 1 (RBFOX1), transcript

348





variant 1, mRNA.



RBFOX1
intronic
NM_145892

Homo sapiens RNA binding protein, fox-1 homolog (C. elegans) 1 (RBFOX1), transcript

349





variant 2, mRNA.



RBFOX1
intronic
NM 145893

Homo sapiens RNA binding protein, fox-1 homolog (C. elegans) 1 (RBFOX1), transcript

350





variant 3, mRNA.



PRKCB
both
NM_002738

Homo sapiens protein kinase C, beta (PRKCB), transcript variant 2, mRNA.

351


PRKCB
both
NM_212535

Homo sapiens protein kinase C, beta (PRKCB), transcript variant 1, mRNA.

352


FUK
both
NM 145059

Homo sapiens fucokinase (FUK), mRNA.

353


COG4
exonic
NM_001195139

Homo sapiens component of oligomeric golgi complex 4 (COG4), transcript variant 2,

354





mRNA.



COG4
exonic
NM_015386

Homo sapiens component of oligomeric golgi complex 4 (COG4), transcript variant 1,

355





mRNA.



HPR
exonic
NM_020995

Homo sapiens haptoglobin-related protein (HPR), mRNA.

356


RPL38
exonic
NM 000999

Homo sapiens ribosomal protein L38 (RPL38), transcript variant 1, mRNA.

357


RPL38
exonic
NM 001035258

Homo sapiens ribosomal protein L38 (RPL38), transcript variant 2, mRNA.

358


MGC16275
exonic
NR 026914

Homo sapiens uncharacterized protein MGC16275 (MGC16275), non-coding RNA.

359


TTYH2
exonic
NM_032646

Homo sapiens tweety family member 2 (TTYH2), transcript variant 1, mRNA.

360


TTYH2
exonic
NM_052869

Homo sapiens tweety family member 2 (TTYH2), transcript variant 2, mRNA.

361


DNAI2
exonic
NM 001172810

Homo sapiens dynein, axonemal, intermediate chain 2 (DNAI2), transcript variant 2, mRNA.

362


DNAI2
exonic
NM 023036

Homo sapiens dynein, axonemal, intermediate chain 2 (DNAI2), transcript variant 1, mRNA.

363


KIF19
exonic
NM_153209

Homo sapiens kinesin family member 19 (KIF19), mRNA.

364


BTBD17
exonic
NM_001080466

Homo sapiens BTB (POZ) domain containing 17 (BTBD17), mRNA.

365


GPR142
exonic
NM_181790

Homo sapiens G protein-coupled receptor 142 (GPR142), mRNA.

366


GPRC5C
exonic
NM_022036

Homo sapiens G protein-coupled receptor, family C, group 5, member C (GPRC5C),

367





transcript variant 1, mRNA.



GPRC5C
exonic
NM 018653

Homo sapiens G protein-coupled receptor, family C, group 5, member C (GPRC5C),

368





transcript variant 2, mRNA.



CD300A
exonic
NM_001256841

Homo sapiens CD300a molecule (CD300A), transcript variant 2, mRNA.

369


CD300A
exonic
NM_007261

Homo sapiens CD300a molecule (CD300A), transcript variant 1, mRNA.

370


CD300LB
exonic
NM 174892

Homo sapiens CD300 molecule-like family member b (CD300LB), mRNA.

371


CD300C
exonic
NM 006678

Homo sapiens CD300c molecule (CD300C), mRNA.

372


CD300LD
exonic
NM 001115152

Homo sapiens CD300 molecule-like family member d (CD300LD), mRNA.

373


C17orf77
exonic
NM_152460

Homo sapiens chromosome 17 open reading frame 77 (C17orf77), mRNA.

374


CD300E
exonic
NM_181449

Homo sapiens CD300e molecule (CD300E), mRNA.

375


RAB37
exonic
NM 175738

Homo sapiens RAB37, member RAS oncogene family (RAB37), transcript variant 3, mRNA.

376


CD300LF
exonic
NM 139018

Homo sapiens CD300 molecule-like family member f (CD300LF), mRNA.

377


RAB37
intronic
NM_001163989

Homo sapiens RAB37, member RAS oncogene family (RAB37), transcript variant 4, mRNA.

378


RAB37
intronic
NM_001006638

Homo sapiens RAB37, member RAS oncogene family (RAB37), transcript variant 2, mRNA.

379


RAB37
intronic
NM_001163990

Homo sapiens RAB37, member RAS oncogene family (RAB37), transcript variant 5, mRNA.

380


TBC1D16
intronic
NM 019020

Homo sapiens TBC1 domain family, member 16 (TBC1D16), transcript variant 1, mRNA.

381


TBC1D16
intronic
NM 001271844

Homo sapiens TBC1 domain family, member 16 (TBC1D16), transcript variant 2, mRNA.

382


TBC1D16
intronic
NM_001271845

Homo sapiens TBC1 domain family, member 16 (TBC1D16), transcript variant 3, mRNA.

383


TBC1D16
intronic
NM_001271846

Homo sapiens TBC1 domain family, member 16 (TBC1D16), transcript variant 4, mRNA.

384


RPTOR
intronic
NM_001163034

Homo sapiens regulatory associated protein of MTOR, complex 1 (RPTOR), transcript

385





variant 2, mRNA.



RPTOR
intronic
NM 020761

Homo sapiens regulatory associated protein of MTOR, complex 1 (RPTOR), transcript

386





variant 1, mRNA.



PTPN2
exonic
NM_001207013

Homo sapiens protein tyrosine phosphatase, non-receptor type 2 (PTPN2), transcript variant 4,

387





mRNA.



PTPN2
exonic
NM_080422

Homo sapiens protein tyrosine phosphatase, non-receptor type 2 (PTPN2), transcript variant 2,

388





mRNA.



PTPN2
exonic
NM 080423

Homo sapiens protein tyrosine phosphatase, non-receptor type 2 (PTPN2), transcript variant 3,

389





mRNA.



PTPN2
intronic
NM 002828

Homo sapiens protein tyrosine phosphatase, non-receptor type 2 (PTPN2), transcript variant 1,

390





mRNA.



ST8SIA5
exonic
NM_013305

Homo sapiens ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 5 (ST8SIA5),

391





mRNA.



PIAS2
exonic
NM 004671

Homo sapiens protein inhibitor of activated STAT, 2 (PIAS2), transcript variant beta, mRNA.

392


PIAS2
exonic
NM_173206

Homo sapiens protein inhibitor of activated STAT, 2 (PIAS2), transcript variant alpha,

393





mRNA.



SERPINB4
exonic
NM_002974

Homo sapiens serpin peptidase inhibitor, clade B (ovalbumin), member 4 (SERPINB4),

394





mRNA.



FLJ26850
intronic
NR_027257

Homo sapiens FLJ26850 protein (FLJ26850), non-coding RNA.

395


FPR2
exonic
NM 001005738

Homo sapiens formyl peptide receptor 2 (FPR2), transcript variant 2, mRNA.

396


FPR2
exonic
NM 001462

Homo sapiens formyl peptide receptor 2 (FPR2), transcript variant 1, mRNA.

397


FPR3
exonic
NM_002030

Homo sapiens formyl peptide receptor 3 (FPR3), mRNA.

398


ZNF577
exonic
NR_024181

Homo sapiens zinc finger protein 577 (ZNF577), transcript variant 3, non-coding RNA.

399


ZNF577
exonic
NM 001135590

Homo sapiens zinc finger protein 577 (ZNF577), transcript variant 2, mRNA.

400


ZNF577
exonic
NM 032679

Homo sapiens zinc finger protein 577 (ZNF577), transcript variant 1, mRNA.

401


ZNF649
exonic
NM 023074

Homo sapiens zinc finger protein 649 (ZNF649), mRNA.

402


ZNF613
exonic
NM_001031721

Homo sapiens zinc finger protein 613 (ZNF613), transcript variant 1, mRNA.

403


ZNF613
exonic
NM_024840

Homo sapiens zinc finger protein 613 (ZNF613), transcript variant 2, mRNA.

404


ZNF350
exonic
NM 021632

Homo sapiens zinc finger protein 350 (ZNF350), mRNA.

405


ZNF615
exonic
NM 001199324

Homo sapiens zinc finger protein 615 (ZNF615), transcript variant 1, mRNA.

406


ZNF615
exonic
NM 198480

Homo sapiens zinc finger protein 615 (ZNF615), transcript variant 2, mRNA.

407


ZNF614
exonic
NM_025040

Homo sapiens zinc finger protein 614 (ZNF614), mRNA.

408


ZNF432
exonic
NM_014650

Homo sapiens zinc finger protein 432 (ZNF432), mRNA.

409


ZNF841
exonic
NM 001136499

Homo sapiens zinc finger protein 841 (ZNF841), mRNA.

410


NLRP12
exonic
NM_001277126

Homo sapiens NLR family, pyrin domain containing 12 (NLRP12), transcript variant 3,

411





mRNA.



NLRP12
exonic
NM_001277129

Homo sapiens NLR family, pyrin domain containing 12 (NLRP12), transcript variant 4,

412





mRNA.



NLRP12
exonic
NM 144687

Homo sapiens NLR family, pyrin domain containing 12 (NLRP12), transcript variant 2,

413





mRNA.



VSTM1
intronic
NM_198481

Homo sapiens V-set and transmembrane domain containing 1 (VSTM1), mRNA.

414


SNX5
exonic
NM_014426

Homo sapiens sorting nexin 5 (SNX5), transcript variant 2, mRNA.

415


SNX5
exonic
NM_152227

Homo sapiens sorting nexin 5 (SNX5), transcript variant 1, mRNA.

416


SNORD17
exonic
NR 003045

Homo sapiens small nucleolar RNA, C/D box 17 (SNORD17), small nucleolar RNA.

417


MGME1
exonic
NM 052865

Homo sapiens mitochondrial genome maintenance exonuclease 1 (MGME1), mRNA.

418


OVOL2
exonic
NM_021220

Homo sapiens ovo-like 2 (Drosophila) (OVOL2), mRNA.

419


ADA
intronic
NM_000022

Homo sapiens adenosine deaminase (ADA), mRNA.

420


NRIP1
exonic
NM_003489

Homo sapiens nuclear receptor interacting protein 1 (NRIP1), mRNA.

421


BACH1
exonic
NR_027655

Homo sapiens BTB and CNC homology 1, basic leucine zipper transcription factor 1

422





(BACH1), transcript variant 3, non-coding RNA.



BACH1
intronic
NM 001186

Homo sapiens BTB and CNC homology 1, basic leucine zipper transcription factor 1

423





(BACH1), transcript variant 2, mRNA.



BACH1
intronic
NM 206866

Homo sapiens BTB and CNC homology 1, basic leucine zipper transcription factor 1

424





(BACH1), transcript variant 1, mRNA.



TRPM2
exonic
NM_003307

Homo sapiens transient receptor potential cation channel, subfamily M, member 2 (TRPM2),

425





transcript variant 1, mRNA.



TRPM2
exonic
NR_038257

Homo sapiens transient receptor potential cation channel, subfamily M, member 2 (TRPM2),

426





transcript variant 2, non-coding RNA.



ADARB1
intronic
NM 001112

Homo sapiens adenosine deaminase, RNA-specific, B1 (ADARB1), transcript variant 1,

427





mRNA.



ADARB1
intronic
NM 001160230

Homo sapiens adenosine deaminase, RNA-specific, B1 (ADARB1), transcript variant 7,

428





mRNA.



ADARB1
intronic
NM_015833

Homo sapiens adenosine deaminase, RNA-specific, B1 (ADARB1), transcript variant 2,

429





mRNA.



ADARB1
intronic
NM_015834

Homo sapiens adenosine deaminase, RNA-specific, B1 (ADARB1), transcript variant 3,

430





mRNA.



ADARB1
intronic
NR 027672

Homo sapiens adenosine deaminase, RNA-specific, B1 (ADARB1), transcript variant 5, non-

431





coding RNA.



ADARB1
intronic
NR 027673

Homo sapiens adenosine deaminase, RNA-specific, B1 (ADARB1), transcript variant 4, non-

432





coding RNA.



ADARB1
intronic
NR_027674

Homo sapiens adenosine deaminase, RNA-specific, B1 (ADARB1), transcript variant 6, non-

433





coding RNA.



ADARB1
intronic
NR_073200

Homo sapiens adenosine deaminase, RNA-specific, B1 (ADARB1), transcript variant 8, non-

434





coding RNA.



APOBEC3A
exonic
NM 001270406

Homo sapiens apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3A

435





(APOBEC3A), transcript variant 3, mRNA.



APOBEC3A
exonic
NM 145699

Homo sapiens apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3A

436





(APOBEC3A), transcript variant 1, mRNA.



APOBEC3A B
intronic
NM 001193289

Homo sapiens APOBEC3A and APOBEC3B deletion hybrid (APOBEC3A B), mRNA.

437


APOBEC3B
exonic
NM_001270411

Homo sapiens apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3B

438





(APOBEC3B), transcript variant 2, mRNA.



APOBEC3B
exonic
NM_004900

Homo sapiens apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3B

439





(APOBEC3B), transcript variant 1, mRNA.



MKL1
intronic
NM_020831

Homo sapiens megakaryoblastic leukemia (translocation) 1 (MKL1), mRNA.

440


TNFRSF13C
exonic
NM_052945

Homo sapiens tumor necrosis factor receptor superfamily, member 13C (TNFRSF13C),

441





mRNA.



CENPM
exonic
NM_001110215

Homo sapiens centromere protein M (CENPM), transcript variant 3, mRNA.

442


CENPM
exonic
NM 001002876

Homo sapiens centromere protein M (CENPM), transcript variant 2, mRNA.

443


CENPM
exonic
NM 024053

Homo sapiens centromere protein M (CENPM), transcript variant 1, mRNA.

444


PPP2R3B
intronic
NM_013239

Homo sapiens protein phosphatase 2, regulatory subunit B″, beta (PPP2R3B), mRNA.

445


VCX
exonic
NM_013452

Homo sapiens variable charge, X-linked (VCX), mRNA.

446


PNPLA4
exonic
NM 004650

Homo sapiens patatin-like phospholipase domain containing 4 (PNPLA4), transcript variant 1,

447





mRNA.



PNPLA4
exonic
NM 001142389

Homo sapiens patatin-like phospholipase domain containing 4 (PNPLA4), transcript variant 2,

448





mRNA.



PNPLA4
exonic
NM_001172672

Homo sapiens patatin-like phospholipase domain containing 4 (PNPLA4), transcript variant 3,

449





mRNA.



MIR651
exonic
NR 030380

Homo sapiens microRNA 651 (MIR651), microRNA.

450


JPX
intronic
NR 024582

Homo sapiens JPX transcript, XIST activator (non-protein coding) (JPX), non-coding RNA.

451


GRIA3
intronic
NM 000828

Homo sapiens glutamate receptor, ionotropic, AMPA 3 (GRIA3), transcript variant 2, mRNA.

452


GRIA3
intronic
NM_007325

Homo sapiens glutamate receptor, ionotropic, AMPA 3 (GRIA3), transcript variant 1, mRNA.

453


GRIA3
intronic
NM_001256743

Homo sapiens glutamate receptor, ionotropic, AMPA 3 (GRIA3), transcript variant 3, mRNA.

454


HMGB3
exonic
NM_005342

Homo sapiens high mobility group box 3 (HMGB3), mRNA.

455









For all genes listed in Table 2 (namely, those relevant to CNV-subregions of interest), Table 4 represents a non-redundant list.









TABLE 5







The set of SNVs reported in Tables 7-10, 14, or 15 that were found in


the 70 PML cases in this study for which WES data were generated.











Chromosome
Position_hg19
REF
ALT
SEQ ID














1
 9777599
C
G
1000


1
 12172008
T
C
1001


1
 24486004
G
T
1002


1
 33476435
C
A
1003


1
 33478900
T
A
1004


1
 33487007
C
T
1005


1
 36932047
C
T
1006


1
 36933715
A
G
1007


1
 42047208
C
G
1008


1
 59248085
G
C
1009


1
 59248339
T
C
1010


1
 92941660
C
T
1011


1
 92946625
G
C
1012


1
 92946625
G
C
1013


1
150053494
C
T
1014


1
155317682
C
T
1015


1
155449630
T
G
1016


1
155450331
C
T
1017


1
182554557
C
T
1018


1
198717250
T
G
1019


1
198717272
A
T
1020


1
206945738
C
T
1021


1
207641950
C
T
1022


1
235840495
G
T
1023


1
235897907
C
T
1024


1
235909815
A
T
1025


2
 24431184
C
T
1026


2
 24432937
C
T
1027


2
 24435599
G
A
1028


2
 47205921
C
T
1029


2
 47273468
A
G
1030


2
 47277182
T
C
1031


2
 55910961
T
C
1032


2
 71337203
C
T
1033


2
 98351032
C
G
1034


2
 98351066
C
T
1035


2
 98351081
C
T
1036


2
113589000
C
T
1037


2
163124051
C
T
1038


2
163133194
T
C
1039


2
163134203
G
T
1040


2
163136505
C
G
1041


2
163139025
C
T
1042


2
163139085
A
T
1043


2
163144899
G
A
1044


2
163174327
C
A
1045


2
163174328
T
G
1046


2
219942026
T
A
1047


2
220023045
C
T
1048


2
230231632
C
T
1049


2
230450646
T
A
1050


3
 38181899
G
T
1051


3
 39323163
A
C
1052


3
 53213691
G
C
1053


3
 53221390
T
C
1054


3
121415370
T
C
1055


3
128204761
C
T
1056


3
128205808
C
T
1057


3
142272098
A
G
1058


3
142274880
G
C
1059


3
142281353
C
G
1060


3
142286928
C
T
1061


3
196199032
A
T
1062


3
196199204
G
T
1063


3
196210704
G
A
1064


3
196210764
T
C
1065


3
196214320
C
T
1066


4
 27019452
C
T
1067


4
 27024170
A
G
1068


4
103522068
A
G
1069


4
103522150
G
A
1070


4
103528328
C
T
1071


4
151199080
G
A
1072


4
151520216
G
A
1073


4
187003729
C
G
1074


4
187004074
C
T
1075


4
187005854
A
C
1076


5
 67591018
A
C
1077


5
 77311370
C
T
1078


5
 77311370
C
T
1079


5
 77334964
T
C
1080


5
 77334964
T
C
1081


5
 77335015
G
T
1082


5
 77335015
G
T
1083


5
 77437092
G
C
1084


5
 77437092
G
C
1085


5
 78596018
G
C
1086


5
138856923
C
T
1087


5
156593120
C
T
1088


5
169081453
G
C
1089


6
 3077139
T
C
1090


6
 12121113
C
T
1091


6
 12122102
T
G
1092


6
 12123538
G
T
1093


6
 12124215
C
T
1094


6
 12125232
C
T
1095


6
 12162068
C
T
1096


6
 12163657
C
T
1097


6
 31928306
A
G
1098


6
 31935750
G
A
1099


6
 31936679
C
T
1100


6
 32797809
C
T
1101


6
 32810794
T
A
1102


6
 32811752
C
T
1103


6
 51483961
T
C
1104


6
 51484077
G
C
1105


6
 51491885
G
A
1106


6
 51497503
C
A
1107


6
 51524339
C
G
1108


6
 51524409
G
T
1109


6
 51612746
G
A
1110


6
 51712759
T
C
1111


6
 51747943
T
A
1112


6
 51798908
C
T
1113


6
 52101833
C
T
1114


6
 83884161
C
G
1115


6
143081232
T
C
1116


6
143092151
T
C
1117


6
143092673
G
A
1118


6
144508353
G
A
1119


6
144508563
G
A
1120


7
 2959240
C
T
1121


7
 2962933
C
T
1122


7
 2983958
T
C
1123


8
 39840234
A
G
1124


8
 39862881
C
T
1125


8
 39862893
T
A
1126


8
 42176189
G
A
1127


8
 48690299
A
G
1128


8
 48773526
T
C
1129


8
 48798507
T
C
1130


8
 48826575
C
G
1131


8
 61654298
T
A
1132


8
 61732632
A
G
1133


8
 61757805
C
T
1134


8
 61769428
A
G
1135


8
 61777914
C
G
1136


8
 61777922
C
G
1137


8
 90990521
T
C
1138


8
100205255
G
A
1139


8
100791158
G
A
1140


8
100865941
G
A
1141


8
145154222
G
A
1142


8
145154222
G
A
1143


8
145154257
C
G
1144


8
145154824
A
C
1145


9
  286491
G
A
1146


9
  286593
C
A
1147


9
  304628
G
A
1148


9
  312134
G
A
1149


9
  328047
T
A
1150


9
  334277
G
A
1151


9
  368128
C
T
1152


9
  399233
A
G
1153


9
  446401
A
G
1154


9
  711359
C
T
1155


9
  713132
G
T
1156


9
 32526077
C
T
1157


9
 32526077
C
T
1158


9
120466814
A
G
1159


9
120475302
A
G
1160


9
120475602
C
T
1161


9
120476568
A
G
1162


9
120476816
C
T
1163


10
 1060218
G
A
1164


10
 14974905
T
C
1165


10
 14976727
G
C
1166


10
 14977469
C
A,T
1167


10
 72358167
G
A
1168


10
 76602923
G
T
1169


10
 76748831
C
T
1170


10
 89720659
G
T
1171


10
 90771767
G
A
1172


10
116045796
G
A
1173


11
 4104626
C
A
1174


11
 4112582
C
T
1175


11
 9598696
G
A
1176


11
 9608330
G
A
1177


11
 36595321
C
T
1178


11
 36596528
G
C
1179


11
 36596863
C
T
1180


11
 36597513
G
A
1181


11
 36614561
G
T
1182


11
 36615033
C
T
1183


11
 67814983
G
A
1184


11
 67818269
G
A
1185


11
 76954833
G
A
1186


11
 76979511
A
G
1187


11
108117787
C
T
1188


11
108119823
T
C
1189


11
108123551
C
T
1190


11
108138003
T
C
1191


11
108143456
C
G
1192


11
108175462
G
A
1193


11
108181014
A
G
1194


11
108186610
G
A
1195


11
108186631
A
G
1196


11
108198384
C
G
1197


11
108202772
G
T
1198


12
 12673965
G
A
1199


12
 12870798
G
A
1200


12
 44166753
A
G
1201


12
 44167821
A
T
1202


12
 64878241
G
A
1203


12
 64879775
C
T
1204


12
 88900891
C
A
1205


12
 93196332
C
T
1206


12
 93205148
T
G
1207


12
112583447
A
C
1208


12
122064788
G
GT
1209


12
133201381
T
A
1210


12
133202816
C
T
1211


12
133209020
G
C
1212


12
133220526
T
C
1213


12
133220544
C
T
1214


12
133237658
T
G
1215


12
133245026
G
A
1216


12
133252406
C
A
1217


12
133253971
C
T
1218


12
133253995
G
A
1219


13
 47466549
T
C
1220


13
108861092
G
T
1221


13
108863591
G
A
1222


14
 21992397
T
C
1223


14
 21993359
G
A
1224


14
 22004996
G
T
1225


14
 24805463
G
T
1226


14
 24806303
G
A
1227


14
 61924007
C
G
1228


14
103369593
G
A
1229


15
 41011016
G
A
1230


15
 68378781
A
C
1231


15
 77329479
C
T
1232


15
 91306241
G
A
1233


15
 91310209
A
G
1234


15
 91326099
C
T
1235


15
 91328219
G
T
1236


15
 91328310
A
G
1237


15
 91341543
A
C
1238


16
 1498408
G
A
1239


16
 1510535
C
T
1240


16
 1524855
C
G
1241


16
 7568296
C
T
1242


16
 7703891
A
G
1243


16
 7714909
C
T
1244


16
 7759119
G
A
1245


16
 7759496
C
T
1246


16
 24124365
A
G
1247


16
 27460020
G
A
1248


16
 30133233
T
C
1249


16
 30134529
A
C
1250


16
 50733536
T
C
1251


16
 50741791
C
T
1252


16
 50741791
C
T
1253


16
 50744688
A
G
1254


16
 50745021
C
T
1255


16
 50753867
G
T
1256


16
 70503095
A
G
1257


16
 81819605
C
T
1258


16
 81902826
C
T
1259


16
 81904539
C
T
1260


16
 81939089
T
C
1261


16
 81942028
C
G
1262


16
 81942175
A
G
1263


16
 81946278
A
G
1264


16
 81960772
C
A
1265


17
 7577069
C
T
1266


17
 16852187
A
G
1267


17
 77926526
C
T
1268


18
 43445580
C
T
1269


18
 43445601
T
G
1270


18
 43456296
C
T
1271


18
 43458306
G
A
1272


18
 43460105
C
A
1273


18
 43464763
C
T
1274


18
 43479473
T
C
1275


18
 43488030
T
C
1276


18
 43496370
G
A
1277


18
 43496539
G
A
1278


18
 43497710
A
G
1279


18
 43523240
C
T
1280


18
 43529551
C
T
1281


18
 43531186
C
T
1282


18
 44392443
T
C
1283


18
 48584504
C
T
1284


18
 56401523
C
T
1285


18
 60036429
G
A
1286


18
 60052034
A
C
1287


19
 4817657
C
T
1288


19
 4817852
G
A
1289


19
 7705818
C
T
1290


19
 7712287
G
C
1291


19
 48631258
G
A
1292


19
 48639022
T
C
1293


20
 3843027
C
A
1294


20
 3846397
C
T
1295


20
 31383307
G
A
1296


20
 31384614
G
T
1297


20
 62305450
C
T
1298


20
 62309621
T
C
1299


20
 62326964
C
G
1300


21
 16338814
T
C
1301


21
 16339852
T
C
1302


21
 30698953
T
G
1303


21
 34809232
C
T
1304


21
 45786650
C
T
1305


21
 45795833
G
T
1306


21
 45795877
G
T
1307


21
 45811411
G
T
1308


21
 45811438
C
T
1309


21
 45815307
T
C
1310


21
 45815331
G
A
1311


21
 45815343
A
G
1312


21
 45815425
C
G
1313


21
 45820196
C
T
1314


21
 45826486
G
A
1315


21
 45826616
C
T
1316


21
 45838333
C
T
1317


21
 45844780
C
T
1318


21
 45845528
G
A
1319


21
 45845661
A
G
1320


21
 45845699
G
A
1321


21
 45855099
C
T
1322


22
 21235389
A
G
1323


22
 23915583
T
C
1324


22
 23915745
G
A
1325


22
 23917192
G
T
1326


22
 36661354
C
T
1327


X
 24759574
G
T
1328


X
 24759574
G
T
1329









Table 5 lists, in order of genomic coordinates, all single nucleotide variants (SNVs) that are relevant to the present study, whether as case-level solutions (Tables 7, 8) or potential solutions (Tables 9, 10), or at the level of variant burden analysis (Tables 14, 15). All genome coordinates are based on hg19.









TABLE 6







Non-redundant list of 419 genes involved in the immune system and/or linked to PML via a CNV











RefSeq Gene
Disease
Gene

Gene Number


Symbol
Model
Source
Source Annotation
(GN)














ACADM
AR
Public_db
MySql
157


ACKR1
AD
Public_db
MySql
158


ACP5
AR
Public_db
PMID: 26052098, 27260006, 27821552
159


ADAR
AD_AR
Public_db
PMID: 26052098, 27260006, 27821552
160


ADARB1
unknown
PBio
PMID: 16227093, 17376196, 19482597, 20220309, 21682836, 21809195, 22001568,
2





22085847, 22113393, 24586166, 24725957, 24760760, 25826567



ADK
AR
PBio
PMID: 17205396, 23592612, 25654762, 25720338, 25979489, 26341819, 26642971
161


AGBL4
unknown
PBio
PMID: 17244818, 21074048, 23085998, 25416787, 25332286, 26502776
3


AICDA
AD_AR
Public_db
MySql; PMID: 23765059
162


AK2
AR
Public_db
PMID: 19043417, 19782549, 20008220, 23765059, 24135998, 24753205, 26454313
163


ALG12
AR
Public_db
MySql
164


ALPL
AD_AR
PBio
PMID: 18821074, 20049532, 20977932, 21191615, 21289095, 23091474, 23454488,
165





23860646, 26219705, 26219711, 26219717



AP3B1
AR
Public_db
MySql; PMID: 11590544, 19782549, 24302998, 24753205, 24916509, 25980904,
166





27889060



AP3B2
AR
Public_db
PMID: 26377319, 27889060
167


AP3D1
AR
Public_db
PMID: 26744459, 27889060
168


APOBEC3A
unknown
PBio
PMID: 16720547, 17303427, 20062055, 20615867, 22896697, 23344558, 23640892,
4





25262471, 25576866, 26416327, 26489798, 26678087



APOBEC3B
unknown
PBio
PMID: 16720547, 17303427, 20062055, 20615867, 22896697, 23344558, 23640892,
6





25262471, 25576866, 26416327, 26489798, 26678087



APOL1
association
Public_db
PMID: 27042682
169


ARHGEF7
unknown
PBio
PMID: 11160719, 16983070, 18378701, 19861492, 25284783, 25500533
8


ASH1L
unknown
PBio
PMID: 17923682, 17981149, 22541069, 24012418, 24244179, 25866973, 26002201,
170





27154821, 27229316, 27434206



ASTN2
unknown
PBio
PMID: 2223091, 8602532, 20573900, 24357807, 24381304, 25146927, 25410587,
9





26514622, 26586575



ATL2
unknown
PBio
PMID: 18270207, 19665976, 25773277
171


ATM
AR
Public_db
MySql; PMID: 19903823, 20301790, 23765059, 24799566, 25692705, 27042682,
172





27484032, 27884168, 27895165



ATR
AD_AR
Public_db
PMID: 17564965, 17151099, 19903823, 20506465, 21615334, 24799566, 25910481
173


AUH
AR
PBio
PMID: 12434311, 12655555, 17130438, 20855850, 25280001, 25597510
10


BACH1
unknown
PBio
PMID: 15068237, 18555605, 22024395, 22791292, 23456643, 23562577, 24752012,
11





25344725, 25391381, 24752012, 26045540, 26894991



BACH2
unknown
PBio
PMID: 17262715, 17991429, 18769450, 22791292, 23728300, 24367030, 24608439,
174





24681888, 24694524, 25123280, 25344725, 25665584, 25686607, 26444573,






26620562, 26731475, 26894991, 26981933



BCL10
AR
Public_db
MySql
176


BDKRB2
unknown
PBio
PMID: 7787759, 18930543, 22047990, 22095814, 24925394
12


BLM
AR
PBio
PMID: 15137905, 15493327, 17210642, 17321898, 19109166, 19709744, 2032252,
177





23572515, 24606147



BLNK
AR
Public_db
PMID: 23765059
178


BLOC1S6
AR
Public_db
MySql
179


BMPR2
AD
PBio
PMID: 15877825, 19191909, 23733693, 24334027
13


C11orf65
unknown
Public_db
MySql
181


C1QA
AR
Public_db
PMID: 27821552
182


C1QB
AR
Public_db
PMID: 27821552
183


C1QC
AR
Public_db
PMID: 27821552
184


C5AR1
unknown
PBio
PMID: 1847994, 22964232, 25041699, 25174320, 25455139, 25539817, 25769922,
185





26059553, 26283482, 26537334



CAPZB
unknown
PBio
PMID: 99354614, 19806181, 22493691, 22706086, 22710966, 22918941, 23178720,
186





26758871



CARD11
AD_AR
Public_db
MySql; PMID: 23765059, 25645939, 26525107
187


CARD9
AR
Public_db
PMID: 27222657
188


CASP8
AR
Public_db
PMID: 22365665, 26454313, 27873163, 27999438
189


CCL11
AD
Public_db
MySql
190


CCL2
association
Public_db
MySql
191


CCL5
association
Public_db
MySql
192


CCR2
association
Public_db
MySql
193


CCR5
association
Public_db
MySql
194


CD180
unknown
PBio
PMID: 9763566, 10880523, 21918197, 21959264, 22484241, 23103284, 23483427,
195





24019553, 25749095, 26371254, 26384474, 26482097, 26555723, 26371254



CD19
AR
Public_db
MySql; PMID: 23765059, 26453379
196


CD209
association
Public_db
MySql
197


CD247
AR
Public_db
PMID: 26454313
198


CD27
AR
Public_db
MySql; PMID: 23765059
199


CD27-AS1
unknown
Public_db
MySql
200


CD300LF
unknown
PBio
PMID: 15184070, 15549731, 17202342, 18688020, 19592130, 22288587, 23072861,
23





23293083, 24035150



CD34
unknown
Public_db
PMID: 27042682
201


CD3D
AR
Public_db
PMID: 23765059, 26454313
202


CD3E
AR
Public_db
PMID: 23765059, 26454313
203


CD3G
AR
Public_db
PMID: 23765059, 26454313
204


CD40
AR
Public_db
MySql; PMID: 23765059, 26453379
205


CD55
unknown
PBio
PMID: 12417446, 1385527, 16406700, 16503113, 17678954, 18424707, 19660813,
207





21143144, 22795896, 24588829, 24639397, 25156074, 25954012, 26423932



CD59
AR
Public_db
MySql
208


CD79A
AR
Public_db
PMID: 23765059
209


CD79B
AR
Public_db
PMID: 23765059
210


CD81
AR
Public_db
MySql
211


CD8A
AR
Public_db
PMID: 26454313
212


CDCA7
AR
Public_db
PMID: 26216346
213


CDKN1B
AD
PBio
PMID: 10799578, 10825149, 10916090, 11123298, 11123306, 15557280, 16410832,
24





17273559, 20854895, 21078910, 22454463, 24317118, 25213837



CEBPB
unknown
Public_db
PMID: 27042682
214


CENPM
unknown
PBio
PMID: 15183305, 16391015, 19711193, 25006165
25


CHD7
AD
Public_db
PMID: 18505430, 18976358, 26454313, 27484032
215


CHEK1
unknown
Public_db
PMID: 19903823, 27042682
216


CIITA
AR
Public_db
PMID: 23765059, 26454313, 27484032
217


CLCN7
AD

PMID: 21107136, 25992615
218


COG4
AR
PBio
PMID: 18086915, 18256213, 20065092, 20143049, 21421995, 23462996, 23865579,
26





24784932, 26125015



COG6
AR
Public_db
MySql
219


COMMD6
unknown
PBio
PMID: 14685242, 15799966, 16573520, 20126548, 25355947, 27441653
27


CORO1A
AR
Public_db
PMID: 23887241, 26454313
220


CR2
AR
Public_db
MySql
221


CRADD
AR
PBio
PMID: 11573962, 21242994, 22323537, 24958727, 26190521, 27135977
28


CRTC3
unknown
PBio
PMID: 15466468, 2032252, 21536665, 23033494, 23241891, 25114223, 25316186,
222





25351958, 26937622



CSF3R
AR
Public_db
PMID: 24753537, 26324699, 27789332
223


CTLA4
AD
Public_db
PMID: 18219311, 25213377, 25329329
224


CTPS1
AR
Public_db
MySql
225


CTSC
AR
Public_db
PMID: 27222657
226


CX3CR1
association
Public_db
MySql
227


CXCL12
association
Public_db
MySql
228


CXCL9
unknown
Public_db
PMID: 27042682
229


CXCR1
association
Public_db
MySql
230


CXCR4
AD
Public_db
PMID: 19782549, 19950235, 23765059, 24753205, 25645939, 28009639
231


CXorf40A
unknown
PBio
PMID: 15541360, 24916366, 26881174
232


CYBB
XLR
Public_db
PMID: 27222657
233


CYP2S1
unknown
PBio
PMID: 15681441, 23933117
234


DCLRE1C
AR
Public_db
PMID: 26454313, 26476407, 27484032
235


DDX1
unknown
Public_db
PMID: 27042682
236


DDX58
AD
Public_db
PMID: 23592984, 25692705, 25794939, 26052098, 26748340, 26848516, 26987611,
237





27260006, 27821552



DHX58
association
Public_db
PMID: 25794939, 26748340, 26848516
238


DKC1
XLR
Public_db
MySql; PMID: 23765059
239


DNER
unknown
PBio
PMID: 15965470, 16298139, 16997755, 17765022, 18474614, 20058045, 20367751,
31





22447725, 23041955, 23328254, 24248099, 24935874, 26869529



DOCK2
AR
Public_db
MySql
241


DSC1
unknown
PBio
PMID: 16048752, 20222919, 22692770, 24460202, 24680560, 25078507, 25244249,
243





26043694, 26758100



DUSP16
unknown
PBio
PMID: 15284860, 21613215, 24311790, 25716993, 26381291, 27162525
32


ECRP
unknown
PBio
PMID: 9826755, 12855582, 25271100, 26184157
33


EDIL3
unknown
PBio
PMID: 22601780, 23518061, 24060278, 24504014, 25385367, 26038125
34


EEA1
unknown
PBio
PMID: 16670179, 22591512, 24491918, 24561067, 26478006, 26909655, 27077111
35


EGR1
unknown
PBio
PMID: 15308739, 19050264, 19812322, 20414733, 21368226, 21622185, 22554935,
244





25613134, 26052046, 26980486, 11910893, 14647476, 18203138, 24627779,






25368162, 27192563



EHF
unknown
PBio
PMID: 16380452, 17027647, 19801549, 20879862, 21617703, 24219556, 25217163
36


ELANE
AD
Public_db
PMID: 20008220, 24145314, 27222657
245


EMB
unknown
PBio
PMID: 8432389, 15917240, 18209069, 19164284, 25773908
37


EPG5
AR
Public_db
MySql; PMID: 21965116, 23222957, 23838600, 26917586, 26927810, 27588602
246


ETF1
unknown
PBio
PMID: 20418372, 25606970, 26384426, 26833392, 27387891
247


ETV6
AD
PBio
PMID: 19264918, 20350664, 21714648, 22438058, 25581430, 25807284, 26718572,
38





27365488



F9
XLR
Public_db
MySql
248


FAS
AD
Public_db
PMID: 27222657
249


FASLG
AD
Public_db
PMID: 27222657
250


FCGR2A
AD_AR
Public_db
MySql
251


FCGR3A
AR
Public_db
MySql
252


FCN3
AR
Public_db
MySql
253


FEZ1
unknown
Public_db
PMID: 27042682
254


FHL2
unknown
PBio
PMID: 16389449, 20592280, 22417706, 22633286, 23212909
39


FOS
unknown
Public_db
PMID: 27042682
255


FOXH1
unknown
Public_db
PMID: 27042682
256


FOXN1
AR
Public_db
MySql
257


FOXP3
XLR
Public_db
PMID: 26454313
258


FPR1
unknown
PBio
PMID: 8994115, 10229829, 10611407, 17084101, 22934745, 23230437, 25605714,
259





25826286, 26101324, 26701131, 27034344, 27100350, 27131862, 27154726



FPR2
unknown
PBio
PMID: 8994115, 10229829, 10611407, 17084101, 22934745, 23230437, 25605714,
41





25826286, 26101324, 26701131, 27034344, 27100350, 27131862, 27154726



FPR3
unknown
PBio
PMID: 8994115, 10229829, 10611407, 17084101, 22934745, 23230437, 25605714,
42





25826286, 26101324, 26701131, 27034344, 27100350, 27131862, 27154726



FUK
unknown
PBio
PMID: 11753075, 12651883, 15774760, 19394435, 19647987, 20363321, 22134107,
43





22203233, 22276660, 22461019, 24239607



G6PC3
AR
Public_db
PMID: 20008220, 24145314, 25879134, 26479985
260


GATA2
AD
Public_db
PMID: 23765059, 23887241
261


GDA
unknown
PBio
PMID: 10595517, 18600524, 20826431, 23838888, 24834013
44


GDPD4
unknown
PBio
PMID: 24373430, 24977479, 24977485, 24977489, 25528375, 25596343
45


GFI1
AD
Public_db
PMID: 20008220, 24145314
262


GOLGB1
unknown
PBio
PMID: 17475246, 21217069, 22034594, 23555793, 24046448
263


GPATCH2
unknown
PBio
PMID: 19432882, 25353171, 25376275
46


GPC5
unknown
PBio
PMID: 24130709, 24943672, 25354479, 26224662, 26349477
47


GPRC5A
unknown
PBio
PMID: 19593893, 20959490, 22239913, 25621293, 25714996, 26165721
264


GRAP2
unknown
Public_db
PMID: 25452106, 25636200, 26246585
265


GRIA3
XLR
PBio
PMID: 10441169, 12682273, 17202328, 18590483, 25904555, 26648591
51


GTPBP4
unknown
PBio
PMID: 17785438, 26015807
52


HAX1
AR
Public_db
PMID: 20008220, 24145314
266


HCN1
AD
PBio
PMID: 9405696, 9630217, 9634236, 9921901, 11133998, 23042740, 23077068,
53





23319474, 24403084, 24747641, 24756635, 25580535, 26578877



HELLS
AR
Public_db
PMID: 26216346
267


HEXA
AR
PBio
PMID: 20301397, 21997228, 23727835, 24445368
54


HIVEP1
association
Public_db
MySql; PMID: 20226436, 26117544
268


HIVEP2
AD
Public_db
MySql; PMID: 21475200, 21936769, 23389689, 24366360, 26153216, 26483320,
269





27003583



HIVEP3
unknown
Public_db
MySql
270


HK2
unknown
PBio
PMID: 2749583, 4265132, 19519254, 2496891, 23874603, 25525876, 25602755
55


HMGB3
unknown
PBio
PMID: 12714519, 15259015, 15358624, 16945912, 22014684, 23994280, 26553261
56


HNRNPLL
unknown
PBio
PMID: 18669861, 18719244, 19100700, 20505149, 22073166, 23934048, 24476532,
271





25825742



HP
unknown
PBio
PMID: 16046400, 19380867, 19795414, 26445729
272


HPCAL1
unknown
PBio
PMID: 12445467, 24699524, 25519916, 26659654, 26729710
273


HPR
unknown
PBio
PMID: 16046400, 19380867, 19795414, 26445729
57


HTR2A
association
Public_db
PMID: 19204164, 24089568, 25078361, 26056932, 27003757, 27042682
274


ICOS
AR
Public_db
MySql; PMID: 19380800, 23765059, 27250108
275


IDI1
unknown
PBio
PMID: 14629038, 17202134, 19454010, 20955688, 22579571, 23585482, 25950736
276


IDI2
unknown
PBio
PMID: 14629038, 17202134, 19454010, 20955688, 22579571, 23585482, 25950736
59


IDI2-AS1
unknown
PBio
See IDI2
60


IDO2
unknown
PBio
PMID: 18219311, 18364004, 19487973, 19799997, 20197554, 20484729, 20693847,
61





21084489, 21406395, 21990421, 22754762, 24391212, 24402311, 24844751,






25477879, 25478733, 25541686, 25949913, 26308414, 27183624



IFIH1
AD
Public_db
PMID: 21156324, 24686847, 24995871, 25794939, 26052098, 26748340, 27260006,
277





27821552



IFNAR1
association
Public_db
PMID: 27821552
278


IFNAR2
AR
Public_db
PMID: 26424569, 27821552
279


IFNG
association
Public_db
MySql
280


IFNGR1
AD_AR
Public_db
MySql
281


IFNGR2
AD_AR
Public_db
MySql; PMID: 15356149, 23161749
282


IFNLR1
unknown
PBio
PMID: 12469119, 12483210, 15166220, 22386267, 22891284, 25634147, 25904743,
62





25941255



IGLL1
AR
Public_db
PMID: 25502423
283


IKBKB
AD_AR
Public_db
MySql; PMID: 17047224, 17072332, 25764117, 25930993, 26117626, 26525107
284


IKBKG
XLD_XLR
Public_db
MySql; PMID: 17047224, 21455173, 21455181, 23765059, 25764117, 25886387,
285





25930993, 26117626, 26525107



IKZF1
AD
Public_db
PMID: 26454313, 26981933
286


IL10
AR
Public_db
PMID: 23887241
287


IL10RA
AR
Public_db
PMID: 23887241
288


IL10RB
AR
Public_db
PMID: 23887241
289


IL12B
AR
Public_db
MySql
290


IL12RB1
AR
Public_db
MySql
291


IL17F
AD
Public_db
PMID: 22284928, 23887241, 24240291, 24690400, 25890879, 27144517
292


IL17RA
AR
Public_db
PMID: 23887241
293


IL1B
AD
Public_db
PMID: 15327898, 20543597, 24248593, 26525107, 27730320, 27873163, 27999438
294


IL21
AR
Public_db
MySql
295


IL21R
AD_AR
Public_db
PMID: 23765059, 23887241
296


IL2RA
AR
Public_db
MySql
297


IL2RG
XLR
Public_db
PMID: 23765059, 26454313, 27484032
298


IL4R
association
Public_db
MySql
299


IL7
unknown
PBio
PMID: 21508983, 22288682, 24507157, 24979548, 25130296, 25214510, 25411246,
300





25734144, 26537673, 26675348, 26908786



IL7R
AR
Public_db
PMID: 26454313, 27484032
301


IRAK4
AD_AR
Public_db
PMID: 23766853, 25232776, 25344726, 25764117, 25886387, 25930993, 26785681,
302





27845762



IRF3
AD
Public_db
PMID: 23388631, 26513235, 26748340
303


IRF7
AR
Public_db
MySql; PMID: 26748340
304


IRF8
AD_AR
Public_db
PMID: 23887241
305


IRGM
association
PBio
PMID: 14707092, 17911638, 22174682, 22722598, 23084913, 23335927
306


ISG15
AR
Public_db
MySql; PMID: 26052098, 27260006, 27821552
307


ITSN2
unknown
PBio
PMID: 11748279, 15020715, 17696400, 17696407, 22558309, 22975684, 23986746,
309





24097067, 24284073, 25797047, 26479042



JAGN1
AR
Public_db
PMID: 25129144
310


JAK3
AR
Public_db
PMID: 23765059, 26454313
311


JMY
unknown
PBio
PMID: 19287377, 20573979, 20574148, 20888769, 21965285, 23291261, 25015719,
312





26223951, 26305109



JPX
association
PBio
PMID: 21029862, 23791181, 23943155
64


JUN
unknown
Public_db
PMID: 16928756, 27042682
313


KANK1
association
PBio
PMID: 18458160, 20164854, 21685469, 24399197, 25961457, 26656975
65


KAT6B
AD
PBio
PMID: 17460191, 17694082, 22715153, 25920810
66


KCTD7
AR
PBio
PMID: 17455289, 20301601, 21710140, 22606975, 22638565, 22748208, 25060828,
67





27629772, 27742667



KITLG
AD
Public_db
PMID: 27042682
314


LAMTOR2
AR
Public_db
MySql; PMID 19782549, 20008220, 24753205
315


LARP4B
unknown
PBio
PMID: 20573744, 23815932, 25534202, 26001795, 26501340, 26644407
69


LCP2
unknown
Public_db
PMID: 12874226, 18219311, 19056831, 23494777, 26246585
317


LIG1
AR
Public_db
MySql
318


LIG4
AR
Public_db
MySql
319


LOC102724297
unknown
Public_db
MySql
320


LOC400710
unknown
PBio
ncRNA, limited gene information; see SNAR gene family (adjacent locus)
321


LRBA
AR
Public_db
MySql; PMID: 23765059, 27873163, 27192563
322


LYST
AR
Public_db
PMID: 19302049, 19782549, 20008220, 24753205, 24916509, 26454313, 27881733
323


MAGEA9
unknown
PBio
PMID: 15222021, 15900605, 21093980, 21791470, 25315972, 25445503, 25755744
324


MAGEA9B
unknown
PBio
PMID: 15222021, 15900605, 21093980, 21791470, 25315972, 25445503, 25755744
325


MAGT1
XLR
Public_db
PMID: 23887241, 27873163, 25504528
326


MALL
unknown
PBio
PMID: 11294831, 19064697, 24101378, 24746959, 26109641, 26622604, 26641089,
72





26772392, 27583248, 27846891



MALT1
AR
Public_db
MySql; PMID: 26525107
327


MAP3K2
unknown
PBio
PMID: 11032806, 11278622, 12138187, 14734742, 16430878, 21333552, 2437584,
328





24847879, 25012295, 26056008



MAPK1
unknown
Public_db
PMID: 14671106, 27042682
329


MAPK3
unknown
Public_db
PMID: 14671106, 27042682
330


MAPK9
unknown
PBio
PMID: 15023353, 23685277, 24673683, 25762148, 26141991
73


MAVS
association
Public_db
PMID: 23582325, 26513235, 26987611
331


MCEE
AR
PBio
PMID: 17846917, 20301409, 21365456, 23726524, 24532006, 25763508, 26725562
74


MECP2
XLD_XLR
Public_db
PMID: 27042682
332


MEX3C
unknown
PBio
PMID: 18779327, 22357625, 22658931, 22863774, 22927639, 23140835, 23446422,
333





23999169, 24706898, 24741071



MGAT5
unknown
PBio
PMID: 12417426, 15585841, 18292539, 20089585, 20117844, 25768892, 26972830
75


MKL1
AR
PBio
PMID: 12944485, 22626970, 26098208, 26098211, 26221020, 26241940, 26405212,
89





26224645, 26554816



MRE11A
AR
Public_db
PMID: 23388631, 23765059
334


MS4A1
AR
Public_db
MySql; PMID: 23765059
335


MSN
unknown
PBio
PMID: 9070665, 10444190, 11777944, 12445265, 14758359, 16368573, 17110458,
336





18025306, 18725395, 21486194, 23526587, 23613524, 24250818, 24358210,






24760896, 25746045



MTHFD1
AR
Both
PMID: 26454313
337


MYD88
AD_AR
Public_db
PMID: 23766853, 25344726, 25764117, 25886387, 25930993, 26371186, 27435819
338


NBN
AD_AR
Public_db
MySql; PMID: 23765059
339


NFIC
unknown
PBio
PMID: 11559801, 15327898, 16928756, 18474555, 19058033, 22205750
340


NFIL3
unknown
PBio
PMID: 20080759, 20697558, 22075207, 23453631, 24070385, 24277151, 24280221,
92





24442434, 24909887, 25092873, 25113970, 25310240, 25611557, 25614966,






25801035, 25993115, 26153760, 26379372, 26806130, 26880402



NFKB1
AD
Public_db
PMID: 22081022, 26279205
341


NFKB2
AD
Public_db
MySql; PMID: 25764117
342


NFKBIA
AD
Public_db
MySql; PMID: 23765059, 25645939, 25764117
343


NHEJ1
AR
Public_db
MySql; PMID: 23765059
344


NLRP12
AD
PBio
PMID: 17947705, 18230725, 20861596, 21978668, 23318142, 23970817, 24282415,
93





24347638, 25249449, 25620184, 25902475, 26083549, 26343520, 26386126, 26521018



NLRP3
AD
Public_db
PMID: 16724804, 19302049, 23592984, 26848516, 27999438
345


NOD2
AD
Public_db
PMID: 16724804, 19302049, 23584365, 26509073, 26848516, 26953272
346


NQO2
unknown
PBio
PMID: 16253210, 16905546, 17720881, 18552348, 26046590
94


NRIP1
unknown
PBio
PMID: 18267075, 23241901, 24969109, 25066731, 25697398, 25879677, 26937622
95


ORAI1
AD_AR
Public_db
PMID: 19075015, 20004786, 21790973, 22144678, 23765059, 26454313, 26469693
347


OSTM1
AR
Public_db
PMID: 16813530, 19507210, 21107136, 23685543
348


OVOL2
AD
PBio
PMID: 16423343, 25267199, 26619963, 26749309
98


PDE3B
unknown
PBio
PMID: 17220874, 23276671, 25816736, 26203135, 26297880, 26374610
99


PDGFRA
association
PBio
PMID: 12660384, 18634583, 18701889, 19246520, 19839938, 20032375, 20569695,
100





21123584, 21975205, 22449623, 22523564, 23771592, 25319708, 25940087



PDSS2
AR
PBio
PMID: 17186472, 18437205, 18784258, 21567994, 21871565, 21983691, 23150520
101


PGM3
AR
Public_db
MySql; PMID: 25502423
349


PHACTR4
unknown
PBio
PMID: 15107502, 17609112, 22215804, 22215812, 22766235, 23076051, 23203801,
102





23319639, 24748504, 26850007



PIAS1
unknown
PBio
PMID: 10805787, 10858346, 14644436, 15297606, 15311277, 17065208, 17540171,
103





18056374, 19857525, 20966256, 22969086, 22982248, 23299081, 24036127



PIAS2
unknown
PBio
PMID: 9724754, 11117529, 12077349, 12764129, 14514699, 15582666, 16460827,
350





19549844, 21156324, 21779164, 22210188, 22982248, 24344134, 25484205,






25434787, 26223632



PIK3CD
AD
Both
MySql; PMID: 24165795, 25133419, 25645939, 26437962, 26453379, 27379089,
104





27426521, 27873163, 14647476, 27192563



PIK3R1
AD_AR
Public_db
PMID: 23765059, 23887241, 25645939, 26246585, 26453379, 27076228, 14647476,
351





27192563



PKHD1
AR
PBio
PMID: 8178487, 15052665, 17450421, 23423256, 24964219, 24984783, 25186187,
105





26502924



PLCG2
AD
Public_db
PMID: 19056831, 23000145, 23765059, 23887241, 25452106, 25636200, 25645939,
352





26246585, 27192563



PMS2
AR
Public_db
MySql; PMID: 23765059
353


PNPLA4
unknown
PBio
PMID: 22289388, 23741432, 26017929, 26164793, 26713677, 26741492, 26968210
107


PNPT1
AR
PBio
PMID: 14563561, 15492272, 16410805, 16687933, 17983748, 19580345, 23084291,
108





23221631, 24143183, 24729470, 25457163



POLA1
XLR
Public_db
PMID: 27019227, 27821552
355


POLE
AR
Public_db
MySql; PMID: 23230001, 23765059, 25948378
356


PPP2R3B
unknown
PBio
PMID: 9847399, 10629059, 11593413, 18353419, 20485545, 26683421
109


PRF1
AD_AR
Public_db
PMID: 17311987, 19302049, 21881043, 24916509, 25776844, 26454313, 26864340,
357





27391055



PRKCB
unknown
PBio
PMID: 10872892, 15488737, 16935002, 17060474, 17395590, 19907441, 21997316,
110





22994860, 23959874, 24550541, 25548371, 24550541, 25808972, 26509731, 26510741



PRKCD
AR
Public_db
MySql; PMID: 23319571, 27250108, 27873163
358


PRKCH
unknown
PBio
PMID: 15327898, 16571806, 18353419, 22114277, 22155788, 22892130, 23868949,
111





24705298, 25617472, 25889880



PRKDC
AD_AR
Public_db
PMID: 12847277, 23722905, 26454313, 26838362, 27980111
359


PROC
AD_AR
PBio
PMID: 2437584, 18751723, 21114396, 22447930, 24162617
360


PSMB8
AR
Public_db
PMID: 26052098, 27260006, 27821552
361


PSTPIP1
AD
PBio
PMID: 9488710, 11313252, 12530983, 14707117, 16724804, 19290936, 19302049,
112





24421327, 25040622, 25645939, 25814341, 26386126, 26919742



PTEN
AD
Public_db
PMID: 26246517, 27426521
362


PTPN2
unknown
PBio
PMID: 11909529, 12359225, 12847239, 19290937, 19825843, 19930043, 20473312,
113





20564182, 20848498, 21220691, 22080861, 22080863, 22671594, 24442435,






24445916, 24608439, 24849651, 24997008, 25548153, 25581833



PTPRC
AR
Public_db
PMID: 26454313
363


PTPRN2
unknown
PBio
PMID: 9714834, 10426369, 11086001, 11086294, 11793386, 15114673, 19361477,
114





23595248, 24988487, 26141787, 26609326



PURA
AD
Public_db
PMID: 27042682
364


RAB27A
AR
Public_db
PMID: 19302049, 20008220, 21881043, 23810987
365


RAB37
unknown
PBio
PMID: 21805469, 22899725, 26931073, 27798165
115


RAB7A
AD

PMID: 25992615, 27588602
366


RABGEF1
unknown
PBio
PMID: 12505986, 15143060, 15235600, 16499958, 16533754, 16605131, 17341663,
367





20829437, 22846990, 23552075, 24569883, 24957337, 25427001, 26567216,






26588713, 27791468



RAC2
AD
Public_db
MySql
368


RAD51
AD
Public_db
PMID: 25310191, 27042682
369


RAG2
AR
Public_db
PMID: 23765059, 23887241, 26454313, 27808398
371


RBCK1
AR
Public_db
MySql; PMID: 21455173, 21455181, 23765059, 23969028, 24958845, 25764117,
372





25930993, 26008899, 26525107, 27810922



RBFOX1
unknown
PBio
PMID: 23350840, 24039908, 25043849, 26500751, 26687839
116


RCC1
unknown
PBio
PMID: 1961752, 18442486, 19060893, 20347844, 23536659, 25452301, 26864624
117


RFX5
AR
Public_db
PMID: 23765059, 26454313
373


RFXANK
AR
Public_db
PMID: 23765059, 26454313
374


RFXAP
AR
Public_db
PMID: 23765059, 26454313
375


RGCC
unknown
PBio
PMID: 19158077, 19652095, 23000427, 24973210, 25770350, 26134570
118


RHOQ
unknown
PBio
PMID: 10490598, 12456725, 14734537, 16246732, 17016434, 19258391, 22916134,
119





24223996, 24297911, 24663214, 24667291



RIPK1
association
Public_db
PMID: 21455173, 27999438
376


RIPK3
association
Public_db
PMID: 22365665, 27999438
377


RMRP
AR
Public_db
MySql; PMID 19782549, 20008220, 24753205
378


RNASE3
unknown
PBio
PMID: 19515815, 26184157
120


RNASEH2A
AR
Public_db
PMID: 26052098, 27260006, 27821552
379


RNASEH2B
AR
Public_db
PMID: 26052098, 27260006, 27821552
380


RNASEH2C
AR
Public_db
PMID: 26052098, 27260006, 27821552
381


RNASEL
association
Public_db
PMID: 24995003, 27525044
382


RNF168
AR
Public_db
MySql; PMID: 23765059
383


RNF31
AR
Public_db
PMID: 21455173, 21455181, 23969028, 24958845, 26008899, 26525107, 27810922
384


RNU4ATAC
AR
Public_db
PMID: 27222657
385


RPTOR
unknown
PBio
PMID: 16959881, 22810227, 23349361, 23812589, 24287405, 24303063, 24671993,
123





24948799, 26678875



RTEL1
AR
Public_db
MySql; PMID: 23329068, 23765059, 24009516, 25607374, 26810774
386


RTEL1-
unknown
Public_db
MySql; PMID: 23329068, 23765059, 25607374
387


TNFRSF6B






SALL2
AR
PBio
PMID: 11734654, 15082782, 18818376, 19076363, 19131967, 21362508, 21689070,
388





21791360, 22074632, 22978642, 23029531, 24040083, 24412933, 24903482,






25360671, 25580951, 25608837, 26181197



SAMHD1
AR
Public_db
PMID: 26052098, 27260006, 27821552
389


SBDS
AR
Public_db
PMID: 20008220, 21062271, 27418648, 27658964
390


SERPINB4
unknown
PBio
PMID: 15203215, 19070595, 21857942, 22451727, 22808225, 24560885, 24635038,
124





25111616, 25133778, 25213322



SERPINB6
AR
PBio
PMID: 14670919, 20451170, 24172014, 24359430
125


SH2D1A
XLR
Public_db
MySql; PMID: 19302049, 23765059, 25744037
391


SHARPIN
unknown
Public_db
PMID: 21455181, 22901541, 23969028, 24958845, 26525107, 26848516, 27810922,
392





27892465



SKIV2L
AR
Public_db
PMID: 27260006, 27821552
393


SLC17A5
AR
PBio
PMID: 14742248, 15006695, 15172005, 16575519, 18399798, 20007460, 20951965,
127





21628664, 22778404, 23760462, 23889254, 25494612, 25855729, 25879139



SLC37A4
AR
Public_db
PMID: 20008220, 20301489
394


SLC3A2
unknown
PBio
PMID: 22588539, 22624878, 23297381, 24491544, 25002078, 26172215, 26439699,
126





26444422



SLC46A1
AR
Public_db
PMID: 26454313
395


SLC8A1
unknown
PBio
PMID: 23224883, 23224887, 23224890, 23224891, 26045217, 26418956, 26775040,
396





26859825, 26924806



SMAD2
unknown
Public_db
PMID: 27042682
397


SMAD3
AD
Public_db
PMID: 27042682
398


SMAD4
AD
Both
PMID: 12202226, 14987161, 16800882, 19420158, 25637015, 25705527, 26454313,
399





27042682



SNAP29
AR
Public_db
PMID: 15968592, 21073448, 27588602
400


SNAR-A1
unknown
PBio
PMID: 25327818, 25447144
401


SNAR-A10
unknown
PBio
PMID: 25327818, 25447144
402


SNAR-A11
unknown
PBio
PMID: 25327818, 25447144
403


SNAR-A12
unknown
PBio
PMID: 25327818, 25447144
404


SNAR-A13
unknown
PBio
PMID: 25327818, 25447144
405


SNAR-A14
unknown
PBio
PMID: 25327818, 25447144
406


SNAR-A2
unknown
PBio
PMID: 25327818, 25447144
407


SNAR-A3
unknown
PBio
PMID: 25327818, 25447144
408


SNAR-A4
unknown
PBio
PMID: 25327818, 25447144
409


SNAR-A5
unknown
PBio
PMID: 25327818, 25447144
410


SNAR-A6
unknown
PBio
PMID: 25327818, 25447144
411


SNAR-A7
unknown
PBio
PMID: 25327818, 25447144
412


SNAR-A8
unknown
PBio
PMID: 25327818, 25447144
413


SNAR-A9
unknown
PBio
PMID: 25327818, 25447144
414


SNAR-B1
unknown
PBio
PMID: 25327818, 25447144
415


SNAR-B2
unknown
PBio
PMID: 25327818, 25447144
416


SNAR-C1
unknown
PBio
PMID: 25327818, 25447144
417


SNAR-C2
unknown
PBio
PMID: 25327818, 25447144
418


SNAR-C3
unknown
PBio
PMID: 25327818, 25447144
419


SNAR-C4
unknown
PBio
PMID: 25327818, 25447144
420


SNAR-C5
unknown
PBio
PMID: 25327818, 25447144
421


SNAR-D
unknown
PBio
PMID: 25327818, 25447144
422


SNAR-E
unknown
PBio
PMID: 25327818, 25447144
423


SNAR-F
unknown
PBio
PMID: 25327818, 25447144
424


SNAR-G1
unknown
PBio
PMID: 25327818, 25447144
425


SNAR-G2
unknown
PBio
PMID: 25327818, 25447144
426


SNAR-H
unknown
PBio
PMID: 25327818, 25447144
427


SNAR-I
unknown
PBio
PMID: 25327818, 25447144
428


SNCA
AD
PBio
PMID: 12406186, 14648159, 16953112, 19115126, 19432400, 19652146, 22209147,
429





23378275, 23771222, 24586351, 24593806, 25092570, 25450953, 25522431,






25635231, 25866630, 26087293, 26272943, 26342897, 26646749



SNHG3
unknown
PBio
PMID: 22308462, 22843687, 26373735
128


SNX10
AR
Public_db
PMID: 22499339, 23123320
430


SNX5
unknown
PBio
PMID: 10600472, 11128621, 14499622, 15133132, 15561769, 16857196, 18596235,
130





21725319, 21903422, 21943487, 23213485, 24820351, 26220253



SOCS2
unknown
PBio
PMID: 19279332, 21403007, 22693634, 22795647, 23455506, 24400794, 26216515,
131





26709655, 26765997, 27071013, 27158906, 27330188, 27338192



SP110
AR
Public_db
MySql
431


SP140
unknown
Public_db
MySql
432


SPINK5
AR
Public_db
PMID: 19683336, 26865388, 27222657, 27905021
433


SQSTM1
AD_AR
Public_db
PMID: 19229298, 27715390
434


SRSF1
unknown
Public_db
PMID: 27042682
435


ST8SIA5
unknown
PBio
PMID: 11089916, 15829700
133


STAT2
AR
Public_db
PMID: 23391734, 26122121, 27821552
437


STAT5B
AR
Public_db
MySql
439


STIM1
AD_AR
Public_db
PMID: 20004786, 21790973, 23765059, 26454313, 26469693
440


STIM2
unknown
PBio
PMID: 20004786, 21790973, 21880262, 22129055, 22477146, 22914293, 25157823,
134





26109647, 26469693



STK4
AR
Public_db
PMID: 19782549, 23765059, 23887241, 24753205, 26029204
441


STX11
AR
Public_db
PMID: 19302049, 21881043, 24916509, 26454313
442


STXBP2
AD_AR
Public_db
PMID: 21881043, 24916509, 25564401, 26454313
443


SYNCRIP
unknown
PBio
PMID: 10734137, 18045242, 19331829, 19232660, 22493061, 22935615, 23679954,
444





23700384, 24844655, 25100733, 26641092



T
AD_AR
PBio
PMID: 11897834, 17438107, 23064415, 23662285, 24253444, 24556085, 25186612,
445





26210634, 26919728



TAP1
AR
Public_db
PMID: 26454313
446


TAP2
AR
Public_db
PMID: 26454313
447


TAPBP
unknown
Public_db
PMID: 26454313
448


TAZ
XLR
Public_db
PMID: 20008220
449


TBC1D16
unknown
PBio
PMID: 16923123, 19077034, 21250943, 23019362, 23485563, 23812537, 24513270,
136





26030178



TBK1
AD
Public_db
PMID: 23887241, 25930993, 26513235, 28049150
450


TBX1
AD
Public_db
PMID: 26454313
451


TCIRG1
AD_AR
Public_db
MySql; PMID 19507210, 19782549, 24753205, 27233968
452


TICAM1
AD_AR
Public_db
PMID: 22105173, 23887241, 25764117, 25930993, 26513235, 28049150
453


TLR3
AD
Public_db
PMID: 23592984, 23887241, 25930993, 26513235, 27810922, 27873163, 27881733
454


TLR4
association
Both
PMID: 12124407, 17893200, 18946062, 19843948, 20521908, 21677132, 22474023,
455





22962435, 23055527, 23890253, 25365308, 25454804, 25930993, 26189680,






26453379, 27881733



TMEM173
AD
Public_db
PMID: 23388631, 25645939, 25692705, 26052098, 27260006, 27801882, 27821552
456


TNF
association
Public_db
MySql; PMID: 27042682
457


TNFAIP3
AD
Public_db
PMID: 23969028, 26642243, 27845235
458


TNFRSF10A
unknown
PBio
PMID: 10889508, 11602752, 11704827, 11777550, 11844843, 12390973, 12694389,
138





14975593, 15007095, 16394652, 16554480, 17671142, 19690337, 20921531



TNFRSF11A
AD_AR
Public_db
PMID: 17088646, 17360404, 18281276, 18606301, 19380800, 19507210, 25102334,
459





25393853, 27003757, 27016605



TNFRSF11B
AR
Public_db
PMID: 19507210, 25102334, 25393853, 27003757
460


TNFRSF13B
AD_AR
Public_db
MySql; PMID: 17467261, 17492055, 18978466, 18981294, 19629655, 20889194,
461





21458042, 22697072, 23765059, 25454804, 25930993, 26727773, 27123465



TNFRSF13C
AR
Both
MySql; PMID: 16769579, 17785824, 18784835, 18813230, 19136305, 19406831,
139





20547827, 20547828, 20817206, 21897850, 22028296, 22030463, 23684423,






24101550, 24953530, 25454804, 25637018, 25724205, 26419927, 26453379,






26600308, 26888554



TNFRSF18
unknown
PBio
PMID: 16439533, 19162554, 19363449, 22017440, 23432692, 24484736, 25738498
140


TNFRSF4
AR
Public_db
MySql
462


TNFRSF8
unknown
PBio
PMID: 10921351, 15990453, 16472805, 18852356, 20141444, 20378007, 21933041,
463





23115213, 23307550, 23654079, 24809535, 25999451



TNFSF11
AR
Public_db
PMID: 17088646, 17360404, 18281276, 18606301, 19507210, 25992615, 27003757
464


TNFSF12
association
Public_db
PMID: 23765059
465


TP53
AD_AR
Public_db
MySql; PMID: 11048806, 11079782, 12009037, 19282432, 26870672
466


TRAF3
AD
Public_db
PMID: 20832341, 23887241, 25764117, 25930993, 28049150
467


TRAF6
unknown
Public_db
PMID: 10215628, 10421844, 25200954, 27808398, 27999438
468


TRAFD1
unknown
PBio
PMID: 16221674, 18849341, 23913580, 25909814, 25992615, 26283173
141


TREX1
AD_AR
Public_db
PMID: 26052098, 27260006, 27821552
469


TRNT1
AR
Public_db
MySql; PMID: 25193871
470


TRPM2
unknown
PBio
PMID: 9806837, 16585058, 18569867, 19411837, 20107186, 25012489, 25049394,
142





25088676, 26300888, 26558786, 26679996, 26942016, 26969190, 27405665, 27872485



TTC7A
AR
Public_db
MySql; PMID: 27873163
471


UBE2N
unknown
PBio
PMID: 21512573, 23159053, 24906799, 25343992, 25503582, 25548215, 26085214,
145





26150489, 26212332, 26518362



UNC119
AD
Public_db
MySql
472


UNC13D
AR
Public_db
PMID: 19302049, 21881043, 24916509, 25564401, 25980904, 26454313
473


UNC93B1
association
Public_db
PMID: 23810987, 23887241, 25930993, 27873163
474


UNG
AR
Public_db
MySql; PMID: 23765059
475


USP18
AR
Public_db
PMID: 27016605, 27325888, 27801882, 27821552
476


USP20
unknown
Public_db
PMID: 27801882
477


VAPA
unknown
PBio
PMID: 9657962, 10523508, 10655491, 11511104, 12931207, 18713837, 23536298,
478





24076421, 24569996, 25015719



VCP
AD
Public_db
PMID: 24248593, 27730320
479


VDAC1
unknown
PBio
PMID: 10620603, 25874870, 26322231, 26542804, 26616244, 26758954, 26878172
480


VPS13B
AR
Public_db
PMID: 20008220, 20301655
481


VPS45
AR
Public_db
MySql; PMID: 23738510, 24145314, 24164830, 26358756
482


VSTM1
unknown
PBio
PMID: 22960280, 23436183, 24205237, 25351446, 25887911, 26760041
147


VWA2
unknown
PBio
PMID: 14506275, 18434322, 21385852, 23443151, 23960233, 26121272
148


WEE1
unknown
Public_db
PMID: 19903823, 25088202, 26598692, 26881506, 27042682
484


WIPF1
AR
Public_db
PMID: 23765059, 26029204, 26453379
485


XIAP
XLD_XLR
Public_db
MySql; PMID: 22365665, 25744037, 26953272
486


YBX1
unknown
Public_db
PMID: 27042682
487


YWHAZ
unknown
Both
PMID: 25894827, 27042682
488


ZAP70
AD_AR
Public_db
PMID: 18219311, 23494777, 23765059, 24164480, 26454313
489


ZBTB24
AR
Public_db
MySql; PMID: 23486536, 23765059, 26851945, 27098601
490









Table 6 is a comprehensive list of 419 exemplary genes (referred to herein as ‘PML-419 genes’ or ‘PML-419 gene list’) interrogated in the present study, along with information related to the inheritance pattern assumed for analysis and the reason for inclusion of the gene. Gene sources for Table 6 (column heading ‘Gene_Source’): 1) nominated on the basis of being linked to immune deficiency, as curated from public databases (indicated by ‘Public_db’) such as PubMed and Clin Var, 2) PBio CNV-identified genes (‘PBio’, see Table 6 column heading ‘Gene_Source’) from a genome-wide array CGH gene discovery study of 71 PML cases, or 3) curated from public databases and identified in PBio's PML gene discovery study (indicated by ‘Both’). A genetic predisposition to PML on the basis of the host's genome was proposed; that is, germline genetic variant(s) in the PML patient's genome, rather than genetic variants that are present in the JC virus, are the cause of the patient's PML (Hatchwell, Front Immunol., 6:216 (2015). Details on the source of the genes in the PML-419 gene list can be found in the following immunodeficiency and immune-related gene sources: Durandy et al., Nat Rev Immunol., 13(7):519-33 (2013); Milner et al., Nat Rev Immunol., 13(9):635-48 (2013); Paciolla et al., Genes Immun., 16(4):239-46 (2015); Hatchwell, Front Immunol., 6:216 (2015); Thijssen et al., Nat Commun., 6:7870 (2015); Chinn et al., Immunol Allergy Clin North Am., 35(4):671-94 (2015); Zhou et al., Nat Genet., 48(1):67-73 (2015); Navabi et al., Allergy Asthma Clin Immunol., 12:27 (2016); and Tsujita et al., J Allergy Clin Immunol. (2016). MySq1′ genes are derived from the ClinVar database. ClinVar was searched using the terms “immune deficiency” and “immunodeficiency.” Entries that described large genomic rearrangements, containing multiple genes, were excluded. A non-redundant list of 125 genes was compiled by combining the output of the two searches and deposited into a MySQL database. NOTE: A subset of these genes are not flagged as ‘MySq1’ if they appeared in one or more of the immune gene review papers noted above. van der Kolk et al., Ann Clin Trans' Neurol.; 3(3):226-32 (2016) was the source of known BAG3 PML gene (see below) and 28 candidate PML genes on the basis of connection to JCV. Van der Kolk et al., cite a method as follows: “the latter was performed by searching for JCV in NCBI, and selecting for genes in humans.” This yielded 30 human genes, 5 of which overlapped with the PML gene list and 2 genes (HLA-DQB1, HLA-DRB1) were excluded because HLA loci are difficult to interpret. The genes ADA, BAG3, BTK, CD40LG, DOCKS, STAT1, WAS, and WIPF1 were derived from Hatchwell, Front Immunol., 6:216 (2015) (see Table 1 for primary references); van der Kolk et al., Ann Clin Transl Neurol., 3(3):226-32 (2016); and Zerbe et al., Clin Infect Dis., 62(8):986-94 (2016). PBio genes are based on CNV studies and a subset overlap the immune review gene lists (annotated as ‘Both’ in column heading ‘Gene_Source’). Tier 1 genes were used as potential solutions for PML cases. Determination of Autosomal Dominant (AD), Autosomal Recessive (AR), X-linked dominant (XLD), or X-linked (XLR) disease model for each gene was derived from the immunodeficiency review papers and/or OMIM annotations. Entries marked ‘association’ denotes variants were found to be associated with an immune-related condition; ‘unknown’ denotes no evidence reported in the literature for an AD or AR model.









TABLE 7







Potential cause of PML in each patient in the study





















Varian










Frequency
Frequency









Details
(Reciprocol)




Ethnic-

Primary
RefSeq Gene

(Ethnic
(Ethnic



Sample_ID
ity
Gender
Disease
Symbol
Variant_Type
specific)
specific)
SEQ_ID





MVGS1116-8a
EUR
F
MS
DOCK8
SNV hom:SNV
  0.499:0.00447

1 in 1,792

1147:1148





(NZ Rx)

het





MVGS1359
EUR
F
MS
IL17F
SNV het
0.00024

1 in 4,170

1114





(NZ Rx)







MVGS1368
EUR
F
MS
IDO2
SNV hom:SNV
 0.508:0.065
1 in 121
1125:1126





(NZ Rx)

het





MVGS540-374b
EUR
M
MS
SHARPIN
SNV hom
0.00217
1 in 461
1142





(NZ Rx)







MVGS540-393b
EUR
F
MS
DOCK8
SNV hom:SNV
0.499:0.00153:0.194

1 in 5,246

1147:1154:1152





(NZ Rx)

het:SNV het





MVGS694-6a
EUR
F
Other
CHD7
SNV het
0.00028

1 in 3,528

1135


MVGS811-13a
EUR
M
HIV
PIK3CD_PIK3CD-
CNV hom
novel
0
  2






AS1






MVGS995-4a
EUR
M
MS
EPG5
SNV hom:SNV
  0.495:0.000251
  1 in 32,224
1279:1273





(NZ Rx)

het





PML01
EUR
F
HIV
ITSN2
SNV hom
0.00183
1 in 547
1028


PML02
EUR
M
Other
IKBKB
SNV het
novel
0
1127


PML03
EUR
F
MS
FPR2
CNV hom
2.23E−06
   1 in 448,833
 140





(NZ Rx)







PML04
EUR
M
HIV
unsolved
n/a
n/a
n/a
n/a


PML05
LAT
M
HIV
TBK1
SNV het
novel
0
1203


PML06
AFR
M
HIV
TICAM1
SNV het
 0.000777

1 in 1,287

1289


PML09
EUR
M
HIV
LIG4
SNV:SNV
0.00399:0.287 
 1 in 3497
1221:1222







comp het





PML10
EUR
F
HIV
TNFRSF11A
SNV het
novel
0
1287


PML12
LAT
F
HIV
BLM
SNV hom
 0.000874

1 in 1,144

1235


PML13
AFR
M
HIV
PLCG2
SNV:SNV
0.00167:0.0187
   1 in 128,105
1261:1263







comp het





PML14
EUR
M
HIV
PLCG2
SNV:SNV
0.00998:0.0159
  1 in 25,259
1261:1263







comp het





PML15
LAT
M
HIV
NOD2
SNV het
novel
0
1255


PML16
AFR
F
HIV
TNFRSF11A
SNV het
novel
0
1287


PML17
EUR
M
HIV
ZAP70
SNV het
0.00009
  1 in 11,110
1035


PML18
EUR
M
HIV
unsolved
n/a
n/a
n/a
n/a


PML19
AFR
M
HIV
ATM
SNV:SNV
0.0479:novel
0
1193:1194







comp het





PML20
AFR
M
HIV
NFKB1
SNV het
0.00173
1 in 577
1069


PML21
EUR
M
HIV
ZAP70
SNV het
 0.0000602
  1 in 16,623
1034


PML22
EUR
M
HIV
unsolved
n/a
n/a
n/a
n/a


PML23
EUR
F
HIV
DCLRE1C
SNV hom
novel
0
1167


PML25
EUR
F
HIV
PLCG2
SNV het
 0.000150

1 in 6,672

1259


PML26
EUR
M
HIV
TRAFD1
SNV hom
 0.000689

1 in 1,451

1208


PML27
EUR
M
HIV
TAP2
SNV hom
0.00837
1 in 120
1101


PML28
EUR
F
MS
TRPM2
SNV hom
novel
0
1311





(NZ Rx)







PML29
AFR
M
HIV
KCTD7_RABGEF1
CNV hom
 0.000387

1 in 2,584

 65


PML30
EUR
M
HIV
TNFRSF11A
SNV het
novel
0
1287


PML31
AFR
F
HIV
DDX58
SNV het
 0.000779

1 in 1,283

1157


PML32
EUR
M
HIV
unsolved
n/a
n/a
n/a
n/a


PML33
EUR
M
HIV
TNFRSF11A
SNV het
novel
0
1287


PML35
EUR
F
HIV
TNFRSF11A
SNV het
novel
0
1287


PML36
AFR
F
HIV
TCIRG1
SNV het
 0.002134
1 in 469
1184


PML37
AFR
M
HIV
GATA2
SNV het
novel
0
1056


PML38
EUR
M
HIV
MALL
CNV hom
3.95E−06
   1 in 253,036
 26


PML39
AFR
M
HIV
unsolved
n/a
n/a
n/a
n/a


PML40
LAT
F
HIV
PNPT1
SNV hom
novel
0
1032


PML41
AFR
M
HIV
ZAP70
SNV het
novel
0
1036


PML43
EUR
M
HIV
PTPRC
SNV hom
novel
0
1020


PML44
EUR
M
HIV
TNFRSF11A
SNV het
novel
0
1287


PML45
EUR
F
Other
CARD11
SNV het
0.0024 
1 in 417
1123


PML46
LAT
M
HIV
EPG5
SNV:SNV
0.0123:0.436
1 in 745
1278:1279







comp het





PML48
EUR
M
HIV
SMAD4
SNV het
 0.000901
  1 in 11,100
1284


PML49
EUR
M
HIV
STIM1
SNV het
novel
0
1174


PML50
AFR
M
HIV
NOD2
SNV het
novel
0
1256


PML51
EUR
M
HIV
TICAM1
SNV het
0.00265
1 in 377
1289


PML52
EUR
F
Other
unsolved
n/a
n/a
n/a
n/a


PML53
EUR
M
Other
GFI1
SNV het
0.00003
  1 in 32,635
1011


PML54
EUR
F
HIV
TNFRSF11A
SNV het
novel
0
1287


PML55
EUR
F
HIV
RTEL1
SNV het
0.00326
1 in 307
1299


PML56
EUR
M
HIV
TNFRSF11A
SNV het
novel
0
1287


PML57
EUR
F
Other
TRAF3
SNV het
0.00093

1 in 1,075

1229


PML58
AFR
M
HIV
DOCK8
SNV:SNV
0.0575:0.478
1 in 146
1146:1147







comp het





PML59
AFR
M
HIV
IFIH1
SNV het
0.00281
1 in 356
1040


PML60
EUR
M
HIV
unsolved
n/a
n/a
n/a
n/a


PML61
AFR
F
HIV
TNFRSF11A
SNV het
novel
0
1287


PML62
AFR
F
HIV
unsolved
n/a
n/a
n/a
n/a


PML63
AFR
M
HIV
PLCG2
SNV het
0.00195
1 in 514
1260


PML64
AFR
M
HIV
PIK3R1
SNV het
novel
0
1077


PML65
AFR
M
HIV
ITSN2
CNV hom
0.00313
1 in 319
 14


PML66
AFR
M
HIV
unsolved
n/a
n/a
n/a
n/a


PML67
EUR
F
MS
unsolved (CNV
n/a
n/a
n/a
n/a





(NZ Rx)
data, no WES










data)






PML68
EUR
F
MS
LRBA
SNV hom
0.00162
1 in 618
1073





(NZ Rx)







PML69
EUR
M
Other
EGR1_ETF1
CNV hom
0.001 

1 in 1,005

 45


PML72
AFR
F
HIV
NOD2
SNV het
 0.004036
1 in 248
1252









Table 7 contains a single genetic solution/explanation that is the potential cause of PML in each patient in the study (71 cases were assessed with genome-wide array CGH and 71 were also assessed by whole exome sequencing), with the exception of 19 ‘unsolved’ cases. Solutions are based on a combination of CNV and SNV variants, connected by SEQ IDs to tables 1, 4 and 5. For homozygous or compound heterozygous variant solutions, expected population frequencies were calculated as follows:

Expected population frequency for variant a(freq p) and variant b(freq q)=pq/4.


For example, PML09 has 2 variants, SEQID 1221 and 1222, with individual frequencies in the normal population of 0.00399, 0.287. The expected frequency in an ethnically-matched normal population for this combination is (0.00399*0.287)*0.25=0.000286283=1/3,497.


The Primary_Disease identifiers in Table 7 are: HIV, infection with human immunodeficiency virus; MS (NZ Rx), multiple sclerosis treated with natalizumab; Other, which includes a variety of disorders/conditions (MVGS694-6a had aplastic anemia, PML02 and PML52 had lymphoma, PML45 and PML 57 had chronic lymphocytic leukemia, PML53 had sarcoidosis, and PML69 is a kidney transplant patient who was on belatacept).


Solutions were considered on the basis of presence of rare variants (CNVs and/or SNVs) in or near genes that are listed in Table 6. Both autosomal recessive (AR) and autosomal dominant (AD) disease models comprise this set of solutions, based on finding homozygous SNVs, homozygous CNVs, compound heterozygous SNVs, or heterozygous SNVs. Nine PML cases in Table 7 were considered ‘unsolved’ on the basis of analyzing both CNV and SNV data, and one case (PML67) was assessed for CNVs only since WES data were unavailable. In some instances, a case was considered unsolved for a best solution (Table 7) but alternate solutions were reported in Table 8 (see below).


For PML cases that had more than one potential solution. In these instances, the ‘best’ solution (Table 7) was determined on the basis of rarity of the genetic variant(s) and the relative strength of the biology for the PML-419 genes (Table 6). Alternate solutions are reported in Table 8. For example, for PML case MVGS1116-8a, three solutions were found, which impacted genes DOCK8, HIVEP2, and RNF168. In this example, DOCK8 compound heterozygous SNVs (Table 7, SNV hom:SNV het) were selected as the best solution because DOCK8 is a known PML gene. In another example, PML case MVGS1359 has IL17F (het SNV) listed as the best solution in Table 7 because it is rarer than alternate solutions for the ATR and STXBP2 genes.


While some PML patients may have multiple genes/variants causing and/or contributing to their PML, in many PML patients only a single gene will be the primary cause analogous to patients diagnosed with primary immunodeficiency disorders. In addition to the alternate solutions reported in Table 8, which are based on SNV genetic findings only, additional alternate solutions based on CNV genetic findings are reported in Table 1.









TABLE 8







Alternate genetic solutions/explanations as the potential cause of PML in the study















Variant Frequency
Frequency




RefSeq Gene

Details (Ethnic
(Reciprocol)



Sample ID
Symbol
Variant_Type
specific)
(Ethnic specific)
SEQ ID





MVGS1116-8a
HIVEP2
SNV het
novel
0
1118


MVGS1116-8a
RNF168
SNV hom:SNV het
 0.469:0.00818

1 in 1,041

1063:1066


MVGS1359
ATR
SNV het
0.00393
1 in 254
1058


MVGS1359
STXBP2
SNV het
0.00501
1 in 199
1291


MVGS540-374b
MKL1
CNV hom
3.99E−08
   1 in 25,081,515
 157


MVGS540-393b
PRKDC
SNV het
0.00097

1 in 1,031

1130


MVGS811-13a
CLCN7
SNV het
0.00028

1 in 3,571

1239


MVGS995-4a
KAT6B
SNV het
0.00003
  1 in 33,357
1169


MVGS995-4a
PRF1
SNV het
0.00243
1 in 412
1168


PML03
CDKN1B
SNV het
0.00003
  1 in 32,209
1200


PML05
ATR
SNV het
novel
0
1061


PML05
NFKB1
SNV het
0.00501
1 in 200
1070


PML06
CHD7
SNV het
0.00797
1 in 125
1136


PML06
DOCK8
SNV hom:SNV het
0.478:0.0313
1 in 267
1147:1152


PML09
RIPK3
SNV het
0.00398
1 in 251
1227


PML10
JUN
SNV het
0.00103
1 in 968
1009


PML10
RAG1
SNV het
0.00039

1 in 2,566

1179


PML12
CARD11
SNV het
novel
0
1122


PML12
PRKDC
SNV het
novel
0
1128


PML13
DOCK8
SNV hom:SNV het
0.478:0.0313
1 in 267
1147:1152


PML13
IRAK4
SNV het
novel
0
1202


PML13
PIK3CD
SNV het
0.00679
1 in 147
1000


PML14
NBN
SNV het
0.0039 
1 in 256
1138


PML14
NFKB1
SNV het
novel
0
1071


PML15
ASH1L
SNV:SNV comp het
novel:0.0019
0
1016:1017


PML15
CHD7
SNV het
0.00176
1 in 568
1133


PML15
HIVEP2
SNV het
novel
0
1116


PML15
STIM1
SNV het
0.00587
1 in 170
1175


PML16
TBK1
SNV het
novel
0
1204


PML16
TLR3
SNV het
0.00136
1 in 738
1076


PML17
APOL1
SNV het
0.0021 
1 in 475
1327


PML18
PKHD1
SNV hom:SNV het
0.498:0.0471
1 in 171
1104:1107


PML19
DOCK8
SNV:SNV comp het
0.0575:0.478 
1 in 146
1146:1147


PML19
IFIH1
SNV het
0.00444
1 in 225
1041


PML20
JUN
SNV het
0.00535
1 in 187
1010


PML21
PRKCH
SNV het
novel
0
1228


PML21
PSTPIP1
SNV het
0.00093

1 in 1,074

1232


PML21
RAG2
SNV het
novel
0
1182


PML22
RIPK3
SNV hom
0.00309
1 in 324
1226


PML22
VPS45
SNV het
0.00114
1 in 878
1014


PML23
NOD2
SNV het
novel
0
1251


PML23
RAG1
SNV het
0.00003
  1 in 33,317
1180


PML28
PKHD1
SNV hom:SNV het
0.498:0.0471
1 in 171
1104:1107


PML28
TNFRSF13B
SNV het
0.00929
1 in 108
1267


PML30
RTEL1
SNV het
 0.000124

1 in 8,068

1300


PML31
AP3B1
SNV het
novel
0
1084


PML31
PRKDC
SNV het
novel
0
1129


PML33
STIM2
SNV het
0.00003
  1 in 32,688
1068


PML33
TLR3
SNV hom:SNV het
 0.413:0.00435

1 in 2,227

1075:1074


PML33
TLR4
SNV hom:SNV hom
0.00283:0.00285 
1 in 354
1161:1160


PML35
PRKCB
SNV het
0.00276
1 in 362
1247


PML36
NOD2
SNV het
0.00871
1 in 115
1254


PML36
PIK3CD
SNV het
0.00679
1 in 147
1000


PML37
AP3B1
SNV het
novel
0
1080


PML37
ATR
SNV het
0.00038

1 in 2,601

1059


PML37
WEE1
SNV het
0.00825
1 in 121
1177


PML38
MYD88
SNV het
novel
0
1051


PML40
MCEE
SNV hom
0.01  
1 in 100
1033


PML41
AP3B1
SNV het
0.00173
1 in 577
1082


PML41
CHD7
SNV het
novel
0
1137


PML41
DOCK8
SNV:SNV comp het
0.0575:0.478 
1 in 146
1146:1147


PML41
POLE
SNV hom
0.00019

1 in 5,203

1219


PML41
RNF168
SNV:SNV comp het
0.412:novel 
0
1063:1062


PML43
DOCK8
SNV hom:SNV het
0.499:novel 
0
1147:1150


PML44
DCLRE1C
SNV hom:SNV hom
0.0287:0.00575
1 in 174
1166:1165


PML44
GFI1
SNV het
0.00708
1 in 141
1012


PML45
POLA1
SNV het
novel
0
1328


PML46
AP3B1
SNV het
0.00587
1 in 170
1082


PML46
IL21R
SNV het
0.00573
1 in 175
1248


PML46
PRKDC
SNV het
0.00017

1 in 5,781

1131


PML48
TNFRSF11A
SNV het
0.00233
1 in 429
1286


PML49
DCLRE1C
SNV hom:SNV hom
0.00575:0.0287 
1 in 174
1166:1165


PML49
PTEN
SNV het
novel
0
1171


PML49
RIPK1
SNV het
0.00090

1 in 1,112

1090


PML50
AP3B1
SNV het
0.00387
1 in 259
1078


PML50
PIAS2
SNV het
0.00357
1 in 280
1283


PML50
STXBP2
SNV het
0.00038

1 in 2,598

1290


PML52
GFI1
SNV het
0.00708
1 in 141
1012


PML53
IL1B
SNV het
novel
0
1037


PML53
STXBP2
SNV het
0.00501
1 in 199
1291


PML54
EPG5
SNV:SNV comp het
0.0638:0.495 
1 in 127
1278:1279


PML54
IFNGR2
SNV het
0.00009
  1 in 11,096
1304


PML54
RAG1
SNV het
0.00003
  1 in 33,352
1178


PML54
RAG2
SNV het
novel
0
1183


PML57
PIAS1
SNV het
novel
0
1231


PML57
PKHD1
SNV hom:SNV het
0.498:0.0471
1 in 171
1104:1107


PML57
SKIV2L
SNV hom:SNV hom:SNV het
0.157:0.214:0.0471
1 in 538
1098:1100:1099


PML58
GFI1
SNV het
0.00144
1 in 693
1012


PML59
IFNLR1
SNV het
novel
0
1002


PML59
NOD2
SNV het
0.00404
1 in 248
1252


PML59
NRIP1
SNV hom
0.00711
1 in 141
1301


PML59
RAD51
SNV het
0.00865
1 in 116
1230


PML60
MAPK3
SNV het
novel
0
1250


PML60
TP53
SNV het
0.00048

1 in 2,085

1266


PML61
GATA2
SNV het
0.00024

1 in 4,139

1057


PML61
PTPRC
SNV hom
novel
0
1019


PML61
TNFRSF8
SNV het
novel
0
1001


PML62
PRKCD
SNV het
novel
0
1054


PML63
HTR2A
SNV hom
0.00519
1 in 193
1220


PML63
MAPK3
SNV het
0.00193
1 in 518
1249


PML64
PLCG2
SNV het
0.00044

1 in 2,276

1264


PML64
WEE1
SNV het
novel
0
1176


PML65
IRAK4
SNV het
0.00118
1 in 850
1201


PML66
PIK3CD
SNV het
0.00679
1 in 147
1000


PML68
RAG1
SNV het
0.00586
1 in 171
1181


PML72
CARD11
SNV het
0.00242
1 in 413
1121


PML72
HIVEP1
SNV hom
0.00164
1 in 610
1092


PML72
IFIH1
SNV het
0.00843
1 in 119
1043









Table 8 contains analogous information to Table 7, with the exception that Ethnicity, Gender and Primary_Disease are not repeated. Table 8 contains alternate genetic solutions/explanations as the potential cause of PML for the patients in the study (71 cases were assessed with genome-wide array CGH and 70 were also assessed by whole exome sequencing). Solutions in Table 8 are also case-level and represent secondary, alternative solutions for the cases listed (using the same criteria used to identify potential solutions reported in Table 7). In other words, for some individuals, more than one reasonable solution was identified and, while those in Table 7 are considered the most likely, those in Table 8 are also potential solutions. It can be appreciated by those skilled in the art that further data on new PML cases, patients with genetic-based immunodeficiency disorders, or functional studies on a given gene may support selection of a Table 8 solution as the ‘best’ single solution (i.e., a current Table 7 solution could be considered instead as a Table 8 solution, and vice versa).









TABLE 9







Pairs of SNVs impacting the same gene
























Variant











Frequency




RefSeq





Amino
Details




Gene
Variant


Ref
Alt
Acid
(Ethnic
SEQ


Sample ID
Symbol
Type
Chromosome
Position
Allele
Allele
Change
specific)
ID NO



















MVGS1359
TTC7A
SNV het
2
47273468
A
G
K252R
0.00684
1030


MVGS1359
TTC7A
SNV het
2
47277182
T
C
S318P
0.00683
1031


MVGS1368
RNF168
SNV het
3
196199204
G
T
P401Q
0.46947
1063


MVGS1368
RNF168
SNV het
3
196210764
T
C
n/a
0.00003
1065


MVGS1368
TLR4
SNV het
9
120475302
A
G
D259G
0.10251
1160


MVGS1368
TLR4
SNV het
9
120475602
C
T
T359I
0.10560
1161


MVGS811-13a
HIVEP1
SNV het
6
12121113
C
T
P362L
0.00024
1091


MVGS811-13a
HIVEP1
SNV het
6
12123538
G
T
K1170N
0.08730
1093


MVGS995-4a
EEA1
SNV het
12
93196332
C
T
E840K
0.01949
1206


MVGS995-4a
EEA1
SNV het
12
93205148
T
G
E702D
0.00003
1207


PML02
RBFOX1
SNV het
16
7759119
G
A
G326S
0.00504
1245


PML02
RBFOX1
SNV het
16
7759496
C
T
P401S
novel
1246


PML04
POLE
SNV het
12
133220526
T
C
N1369S
0.22363
1213


PML04
POLE
SNV het
12
133237658
T
G
Q766P
novel
1215


PML05
TLR4
SNV het
9
120475302
A
G
D259G
0.04628
1160


PML05
TLR4
SNV het
9
120475602
C
T
T359I
0.04180
1161


PML05
POLE
SNV het
12
133220526
T
C
N1369S
0.12669
1213


PML05
POLE
SNV het
12
133252406
C
A
A121S
novel
1217


PML10
TLR4
SNV het
9
120475302
A
G
D259G
0.10251
1160


PML10
TLR4
SNV het
9
120475602
C
T
T359I
0.10560
1161


PML12
IDO2
SNV het
8
39840234
A
G
I127V
0.38971
1124


PML12
IDO2
SNV het
8
39862881
C
T
R235W
0.50282
1125


PML12
IDO2
SNV het
8
39862893
T
A
S239T
0.02384
1126


PML13
STX11
SNV het
6
144508353
G
A
V197M
novel
1119


PML13
STX11
SNV het
6
144508563
G
A
V267M
0.00202
1120


PML13
DCLRE1C
SNV het
10
14974905
T
C
H123R
0.16298
1165


PML13
DCLRE1C
SNV het
10
14976727
G
C
P171R
0.22295
1166


PML13
EPG5
SNV het
18
43497710
A
G
V1058A
0.42740
1279


PML13
EPG5
SNV het
18
43531186
C
T
S424N
0.00600
1282


PML14
ATM
SNV het
11
108117787
C
T
S333F
0.00280
1188


PML14
ATM
SNV het
11
108175462
G
A
D1853N
0.24654
1193


PML14
TRPM2
SNV het
21
45815425
C
G
I621M
novel
1313


PML14
TRPM2
SNV het
21
45845699
G
A
V1242M
0.00537
1321


PML16
TLR3
SNV het
4
187004074
C
T
L135F
0.12378
1075


PML16
TLR3
SNV het
4
187005854
A
C
I571L
0.00136
1076


PML16
HIVEP1
SNV het
6
12121113
C
T
P362L
0.07856
1091


PML16
HIVEP1
SNV het
6
12162068
C
T
S160F
0.01979
1096


PML16
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.50029
1104


PML16
PKHD1
SNV het
6
51747943
T
A
D2433V
0.07153
1112


PML16
POLE
SNV het
12
133209020
G
C
Q2044E
novel
1212


PML16
POLE
SNV het
12
133220526
T
C
N1369S
0.24889
1213


PML17
RNF168
SNV het
3
196199204
G
T
P401Q
0.46947
1063


PML17
RNF168
SNV het
3
196210704
G
A
P206L
0.00003
1064


PML17
HIVEP1
SNV het
6
12123538
G
T
K1170N
0.08730
1093


PML17
HIVEP1
SNV het
6
12125232
C
T
S1735F
0.00027
1095


PML17
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.49837
1104


PML17
PKHD1
SNV het
6
51497503
C
A
R3842L
0.04707
1107


PML17
DCLRE1C
SNV het
10
14974905
T
C
H123R
0.27332
1165


PML17
DCLRE1C
SNV het
10
14976727
G
C
P171R
0.13896
1166


PML17
ATM
SNV het
11
108119823
T
C
V410A
0.00643
1189


PML17
ATM
SNV het
11
108175462
G
A
D1853N
0.24654
1193


PML17
EPG5
SNV het
18
43464763
C
T
G1708D
0.00013
1274


PML17
EPG5
SNV het
18
43497710
A
G
V1058A
0.49513
1279


PML18
TLR4
SNV het
9
120475302
A
G
D259G
0.10251
1160


PML18
TLR4
SNV het
9
120475602
C
T
T359I
0.10560
1161


PML20
AK2
SNV het
1
33476435
C
A
n/a
novel
1003


PML20
AK2
SNV het
1
33478900
T
A
Y159F
0.04954
1004


PML20
HIVEP1
SNV het
6
12124215
C
T
P1396L
0.06774
1094


PML20
HIVEP1
SNV het
6
12163657
C
T
P2374S
0.06733
1097


PML20
KANK1
SNV het
9
711359
C
T
S198F
0.11985
1155


PML20
KANK1
SNV het
9
713132
G
T
G631V
0.00136
1156


PML21
DOCK8
SNV het
9
286593
C
A
P29T
0.49889
1147


PML21
DOCK8
SNV het
9
286593
C
A
P29T
0.49889
1147


PML21
DOCK8
SNV het
9
312134
G
A
E169K
0.06358
1149


PML21
DOCK8
SNV het
9
312134
G
A
E169K
0.06358
1149


PML21
TLR4
SNV het
9
120475302
A
G
D259G
0.10251
1160


PML21
TLR4
SNV het
9
120475302
A
G
D259G
0.10251
1160


PML21
TLR4
SNV het
9
120475602
C
T
T359I
0.10560
1161


PML21
TLR4
SNV het
9
120475602
C
T
T359I
0.10560
1161


PML21
ATM
SNV het
11
108138003
T
C
F858L
0.02864
1191


PML21
ATM
SNV het
11
108138003
T
C
F858L
0.02864
1191


PML21
ATM
SNV het
11
108143456
C
G
P1054R
0.05069
1192


PML21
ATM
SNV het
11
108143456
C
G
P1054R
0.05069
1192


PML21
TRPM2
SNV het
21
45786650
C
T
S146F
0.00072
1305


PML21
TRPM2
SNV het
21
45786650
C
T
S146F
0.00072
1305


PML21
TRPM2
SNV het
21
45820196
C
T
R735C
0.10374
1314


PML21
TRPM2
SNV het
21
45820196
C
T
R735C
0.10374
1314


PML22
SKIV2L
SNV het
6
31928306
A
G
Q151R
0.15759
1098


PML22
SKIV2L
SNV het
6
31935750
G
A
V724M
0.04718
1099


PML22
SKIV2L
SNV het
6
31936679
C
T
A1071V
0.21419
1100


PML22
DOCK8
SNV het
9
286593
C
A
P29T
0.49889
1147


PML22
DOCK8
SNV het
9
304628
G
A
R151Q
0.00447
1148


PML22
GDPD4
SNV het
11
76954833
G
A
H383Y
0.44867
1186


PML22
GDPD4
SNV het
11
76979511
A
G
I233T
0.00504
1187


PML22
ATM
SNV het
11
108117787
C
T
S333F
0.00280
1188


PML22
ATM
SNV het
11
108175462
G
A
D1853N
0.24654
1193


PML22
BLM
SNV het
15
91306241
G
A
R643H
0.00799
1233


PML22
BLM
SNV het
15
91341543
A
C
N1112H
novel
1238


PML23
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.49837
1104


PML23
PKHD1
SNV het
6
51497503
C
A
R3842L
0.04707
1107


PML23
SHARPIN
SNV het
8
145154222
G
A
P294S
0.08789
1142


PML23
SHARPIN
SNV het
8
145154257
C
G
S282T
0.14880
1144


PML23
DOCK8
SNV het
9
286491
G
A
D63N
0.27362
1146


PML23
DOCK8
SNV het
9
334277
G
A
R325H
0.00015
1151


PML25
SKIV2L
SNV het
6
31928306
A
G
Q151R
0.15759
1098


PML25
SKIV2L
SNV het
6
31935750
G
A
V724M
0.04718
1099


PML25
SKIV2L
SNV het
6
31936679
C
T
A1071V
0.21419
1100


PML25
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.49837
1104


PML25
PKHD1
SNV het
6
51524409
G
T
S3505R
0.02049
1109


PML25
EPG5
SNV het
18
43445601
T
G
I174L
novel
1270


PML25
EPG5
SNV het
18
43531186
C
T
S424N
0.02391
1282


PML27
LYST
SNV het
1
235897907
C
T
G2804D
0.00114
1024


PML27
LYST
SNV het
1
235909815
A
T
F165Y
0.00102
1025


PML27
EPG5
SNV het
18
43445601
T
G
I174L
novel
1270


PML27
EPG5
SNV het
18
43497710
A
G
V1058A
0.49513
1279


PML29
LIG1
SNV het
19
48631258
G
A
T546I
0.07515
1292


PML29
LIG1
SNV het
19
48639022
T
C
M412V
0.05385
1293


PML30
DCLRE1C
SNV het
10
14974905
T
C
H123R
0.27332
1165


PML30
DCLRE1C
SNV het
10
14976727
G
C
P171R
0.13896
1166


PML30
ATM
SNV het
11
108138003
T
C
F858L
0.02864
1191


PML30
ATM
SNV het
11
108143456
C
G
P1054R
0.05069
1192


PML30
ATM
SNV het
11
108186610
G
A
G2023R
0.00465
1195


PML31
LYST
SNV het
1
235897907
C
T
G2804D
0.23000
1024


PML31
LYST
SNV het
1
235909815
A
T
F165Y
0.15155
1025


PML31
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.50029
1104


PML31
PKHD1
SNV het
6
51524339
C
G
E3529Q
0.07244
1108


PML31
PKHD1
SNV het
6
51747943
T
A
D2433V
0.07153
1112


PML31
PKHD1
SNV het
6
51798908
C
T
G2041S
0.00173
1113


PML32
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.49837
1104


PML32
PKHD1
SNV het
6
51491885
G
A
Q3899*
novel
1106


PML32
EPG5
SNV het
18
43496539
G
A
S1083L
0.06375
1278


PML32
EPG5
SNV het
18
43497710
A
G
V1058A
0.49513
1279


PML32
EPG5
SNV het
18
43529551
C
T
V466M
0.00006
1281


PML33
AK2
SNV het
1
33476435
C
A
n/a
novel
1003


PML33
AK2
SNV het
1
33487007
C
T
S129N
0.01100
1005


PML33
EPG5
SNV het
18
43497710
A
G
V1058A
0.49513
1279


PML33
EPG5
SNV het
18
43523240
C
T
M610I
0.00066
1280


PML35
RNF168
SNV het
3
196199204
G
T
P401Q
0.46947
1063


PML35
RNF168
SNV het
3
196214320
C
T
E170K
0.00818
1066


PML36
IDO2
SNV het
8
39862881
C
T
R235W
0.46108
1125


PML36
IDO2
SNV het
8
39862893
T
A
S239T
0.01135
1126


PML39
LYST
SNV het
1
235897907
C
T
G2804D
0.23000
1024


PML39
LYST
SNV het
1
235909815
A
T
F165Y
0.15155
1025


PML39
NHEJ1
SNV het
2
219942026
T
A
Q181L
0.06324
1047


PML39
NHEJ1
SNV het
2
220023045
C
T
A14T
0.23543
1048


PML40
ATM
SNV het
11
108186631
A
G
I2030V
0.00173
1196


PML40
ATM
SNV het
11
108186631
A
G
I2030V
0.03446
1196


PML40
ATM
SNV het
11
108198384
C
G
L2330V
0.00035
1197


PML40
ATM
SNV het
11
108198384
C
G
L2330V
0.00491
1197


PML41
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.50029
1104


PML41
PKHD1
SNV het
6
51497503
C
A
R3842L
0.00654
1107


PML41
IDO2
SNV het
8
39840234
A
G
I127V
0.06350
1124


PML41
IDO2
SNV het
8
39862881
C
T
R235W
0.46108
1125


PML45
VPS13B
SNV het
8
100791158
G
A
E2560K
0.00964
1140


PML45
VPS13B
SNV het
8
100865941
G
A
A3442T
novel
1141


PML48
EPG5
SNV het
18
43497710
A
G
V1058A
0.49513
1279


PML48
EPG5
SNV het
18
43531186
C
T
S424N
0.02391
1282


PML51
TRPM2
SNV het
21
45826486
G
A
V914I
novel
1315


PML51
TRPM2
SNV het
21
45855099
C
T
R1300W
0.00021
1322


PML53
EPG5
SNV het
18
43445580
C
T
D181N
novel
1269


PML53
EPG5
SNV het
18
43497710
A
G
V1058A
0.49513
1279


PML56
TLR4
SNV het
9
120475302
A
G
D259G
0.10251
1160


PML56
TLR4
SNV het
9
120475602
C
T
T359I
0.10560
1161


PML56
DCLRE1C
SNV het
10
14974905
T
C
H123R
0.27332
1165


PML56
DCLRE1C
SNV het
10
14976727
G
C
P171R
0.13896
1166


PML57
CSF3R
SNV het
1
36932047
C
T
E359K
0.01706
1006


PML57
CSF3R
SNV het
1
36933715
A
G
Y113H
0.00087
1007


PML57
TLR4
SNV het
9
120475302
A
G
D259G
0.10251
1160


PML57
TLR4
SNV het
9
120475602
C
T
T359I
0.10560
1161


PML57
ATM
SNV het
11
108138003
T
C
F858L
0.02864
1191


PML57
ATM
SNV het
11
108143456
C
G
P1054R
0.05069
1192


PML57
ATM
SNV het
11
108175462
G
A
D1853N
0.24654
1193


PML58
DOCK8
SNV het
9
399233
A
G
N1002D
0.19737
1153


PML58
DCLRE1C
SNV het
10
14974905
T
C
H123R
0.16298
1165


PML58
DCLRE1C
SNV het
10
14976727
G
C
P171R
0.22295
1166


PML58
DNMT3B
SNV het
20
31383307
G
A
G311S
0.00192
1296


PML58
DNMT3B
SNV het
20
31384614
G
T
G343V
novel
1297


PML59
LYST
SNV het
1
235897907
C
T
G2804D
0.23000
1024


PML59
LYST
SNV het
1
235897907
C
T
G2804D
0.23000
1024


PML59
LYST
SNV het
1
235909815
A
T
F165Y
0.15155
1025


PML59
LYST
SNV het
1
235909815
A
T
F165Y
0.15155
1025


PML59
LIG1
SNV het
19
48631258
G
A
T546I
0.07515
1292


PML59
LIG1
SNV het
19
48631258
G
A
T546I
0.07515
1292


PML59
LIG1
SNV het
19
48639022
T
C
M412V
0.05385
1293


PML59
LIG1
SNV het
19
48639022
T
C
M412V
0.05385
1293


PML60
DCLRE1C
SNV het
10
14974905
T
C
H123R
0.27332
1165


PML60
DCLRE1C
SNV het
10
14976727
G
C
P171R
0.13896
1166


PML60
POLE
SNV het
12
133202816
C
T
E2113K
0.04686
1211


PML60
POLE
SNV het
12
133220526
T
C
N1369S
0.22363
1213


PML62
TLR4
SNV het
9
120475302
A
G
D259G
0.13066
1160


PML62
TLR4
SNV het
9
120475602
C
T
T359I
0.02672
1161


PML63
HIVEP1
SNV het
6
12124215
C
T
P1396L
0.06774
1094


PML63
HIVEP1
SNV het
6
12163657
C
T
P2374S
0.06733
1097


PML63
PLCG2
SNV het
16
81942175
A
G
N571S
0.01870
1263


PML63
TRPM2
SNV het
21
45795833
G
T
V297L
0.00097
1306


PML63
TRPM2
SNV het
21
45815307
T
C
V582A
0.00724
1310


PML64
DNER
SNV het
2
230231632
C
T
D687N
0.00058
1049


PML64
DNER
SNV het
2
230450646
T
A
T259S
0.00692
1050


PML64
IDO2
SNV het
8
39862881
C
T
R235W
0.46108
1125


PML64
IDO2
SNV het
8
39862893
T
A
S239T
0.01135
1126


PML65
POLE
SNV het
12
133201381
T
A
I2228F
0.00232
1210


PML65
POLE
SNV het
12
133253971
C
T
R233Q
0.02037
1218


PML66
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.50029
1104


PML66
PKHD1
SNV het
6
51612746
G
A
S3223L
0.00000
1110


PML66
PKHD1
SNV het
6
51712759
T
C
T2641A
0.04812
1111


PML66
EPG5
SNV het
18
43456296
C
T
R1985Q
0.07733
1271


PML66
EPG5
SNV het
18
43497710
A
G
V1058A
0.42740
1279


PML68
DCLRE1C
SNV het
10
14974905
T
C
H123R
0.27332
1165


PML68
DCLRE1C
SNV het
10
14976727
G
C
P171R
0.13896
1166


PML72
PSMB8
SNV het
6
32810794
T
A
T70S
0.04224
1102


PML72
PSMB8
SNV het
6
32811752
C
T
G8R
0.04845
1103


PML72
POLE
SNV het
12
133220526
T
C
N1369S
0.24889
1213


PML72
POLE
SNV het
12
133245026
G
A
P477S
0.02332
1216


PML72
RBFOX1
SNV het
16
7568296
C
T
P102S
0.00692
1242


PML72
RBFOX1
SNV het
16
7703891
A
G
T235A
novel
1243









Table 9 lists, for each case (in multiple rows), variants for which it was not possible, using the whole exome sequencing (WES) data available, to determine phase (i.e., whether two variants are in cis—on the same chromosome—or trans—on opposite chromosomes). Determining phase is an important consideration when dealing with disorders that are being evaluated on an autosomal recessive (AR) basis. If two variants are known to be present but it is impossible to determine whether they are in cis or trans, then it is impossible to conclude that both gene copies are affected, as opposed to only one (albeit with 2 variants). This problem does not arise in the case of homozygous variants, for which it is obvious that the variants must be in trans (i.e., it is only an issue for non-identical variants). All genome coordinates are based on hg19 build.


In summary, Table 9 lists all unphased case-level compound heterozygous SNV solutions, which might represent further case-level solutions, were phasing to have been possible. Furthermore, it can be appreciated by those skilled in the art that unphased solutions reported in Table 9 (2 het SNVs per gene) or Table 10 (see below, which reports het SNVs in patients that also have a CNV reported in Table 1) can potentially cause or contribute to the patient's PML if follow up genetic analysis reveals the pair of variants are on different alleles (i.e., each gene copy impacted by a variant). Variants reported in Tables 1, 9, or 10 may also be found to be significantly deleterious on their own (e.g., in functional studies on patient-derived cells, animal models, etc.) and thus constitute an AD model solution (i.e., genes presently listed as ‘AR’ model in Table 6) may be causal or contributing to disease via an AD or AR model, like several genes already known to be AD or AR (Table 6, ‘AD_AR’ disease model).









TABLE 10







SNVs found in genes suspected of being impacted by acquired CNVs
























Variant











Frequency




RefSeq





Amino
Details




Gene
Variant


Ref
Alt
Acid
(Ethnic
SEQ


Sample_ID
Symbol
Type
Chr
Position
Allele
Allele
Change
specific)
ID NO



















MVGS811-13a
NRIP1
SNV het
21
16338814
T
C
N567S
0.00060
1301


MVGS995-4a
VWA2
SNV het
10
116045796
G
A
V366M
0.02392
1173


PML01
PKHD1
SNV het
6
51497503
C
A
R3842L
0.04707
1107


PML01
PKHD1
SNV het
6
51483961
T
C
Q4048R
0.49837
1104


PML02
DUSP16
SNV het
12
12673965
G
A
T23M
0.00015
1199


PML39
SALL2
SNV het
14
22004996
G
T
S13R
0.00231
1225


PML51
JMY
SNV het
5
78596018
G
C
D524H
novel
1086


PML65
SALL2
SNV het
14
21992397
T
C
S347G
0.07709
1223





NOTE:


These are het SNVs that are potentially compound heterozygotes with a CNV on the allele. See text for description.


The DUSP16 SNV (chr12: 12673965) was in trans with a chr12 deletion of DUSP16 in this patient (PML02), whose primary diagnosis was lymphoma.






Table 10 is a list of all heterozygous SNVs that are potentially compound heterozygotes with a CNV on the allele. See text for a fuller explanation. All genome coordinates are based on hg19 build.









TABLE 11







Key that maps Sample_ID for the PML cases


to the PML_Case_ID numbers










Sample_ID
PML_Case_ID







MVGS1116-8a
3006



MVGS1359
3117



MVGS1368
3118



MVGS540-374b
3005



MVGS540-393b
3004



MVGS694-6a
3007



MVGS811-13a
3009



MVGS995-4a
3010



PML01
3127



PML02
3126



PML03
3155



PML04
3156



PML05
3125



PML06
3124



PML09
3132



PML10
3157



PML12
3159



PML13
3160



PML14
3161



PML15
3194



PML16
3163



PML17
3140



PML18
3141



PML19
3164



PML20
3143



PML21
3144



PML22
3145



PML23
3165



PML25
3166



PML26
3167



PML27
3168



PML28
3151



PML29
3152



PML30
3153



PML31
3154



PML32
3169



PML33
3170



PML35
3171



PML36
3172



PML37
3173



PML38
3174



PML39
3175



PML40
3273



PML41
3177



PML43
3178



PML44
3179



PML45
3180



PML46
3196



PML48
3197



PML49
3183



PML50
3198



PML51
3185



PML52
3186



PML53
3187



PML54
3188



PML55
3189



PML56
3190



PML57
3191



PML58
3192



PML59
3193



PML60
3199



PML61
3200



PML62
3201



PML63
3202



PML64
3203



PML65
3204



PML66
3205



PML67
3277



PML68
3278



PML69
3279



PML72
3282



PML70_control
3280



PML71_control
3281



PML73_control
3283



PML74_control
3284



PML75_control
3285



PML76_control
3286










Table 11 provides the Sample_ID and PML_Case_ID (experimental ID for CGH data) for 77 ‘PML cases’ (includes 6 non-PML HIV cases listed as controls).









TABLE 12







Non-redundant list of transcript variants that correspond to the set of genes that no CNV ‘solutions’ have been reported in the 71 PML cases










RefSeq_Gene_Symbol
RefSeq_Accession_Number
mRNA_Description
SEQ ID





ACADM
NM_000016

Homo sapiens acyl-CoA dehydrogenase, C-4 to C-12 straight chain (ACADM), transcript variant 1, mRNA.

1500


ACADM
NM_001127328

Homo sapiens atypical chemokine receptor 1 (Duffy blood group) (ACKR1), transcript variant 1, mRNA.

1501


ACKR1
NM_002036

Homo sapiens atypical chemokine receptor 1 (Duffy blood group) (ACKR1), transcript variant 2, mRNA.

1502


ACKR1
NM_001122951

Homo sapiens atypical chemokine receptor 1 isoform a

1503


ACP5
NM_001611

Homo sapiens acid phosphatase 5, tartrate resistant (ACP5), transcript variant 4, mRNA.

1504


ACP5
NM_001111034

Homo sapiens acid phosphatase 5, tartrate resistant (ACP5), transcript variant 2, mRNA.

1505


ACP5
NM_001111035

Homo sapiens acid phosphatase 5, tartrate resistant (ACP5), transcript variant 1, mRNA.

1506


ACP5
NM_001111036

Homo sapiens acid phosphatase 5, tartrate resistant (ACP5), transcript variant 3, mRNA.

1507


ADAR
NM_001111

Homo sapiens adenosine deaminase, RNA-specific (ADAR), transcript variant 1, mRNA.

1508


ADAR
NM_015840

Homo sapiens adenosine deaminase, RNA-specific (ADAR), transcript variant 2, mRNA.

1509


ADAR
NM_015841

Homo sapiens adenosine deaminase, RNA-specific (ADAR), transcript variant 3, mRNA.

1510


ADAR
NM_001025107

Homo sapiens adenosine deaminase, RNA-specific (ADAR), transcript variant 4, mRNA.

1511


ADAR
NM_001193495

Homo sapiens adenosine deaminase, RNA-specific (ADAR), transcript variant 5, mRNA.

1512


ADK
NM_001202450

Homo sapiens adenosine kinase (ADK), transcript variant 4, mRNA.

1513


ADK
NM_006721

Homo sapiens adenosine kinase (ADK), transcript variant 2, mRNA.

1514


ADK
NM_001123

Homo sapiens adenosine kinase (ADK), transcript variant 1, mRNA.

1515


ADK
NM_001202449

Homo sapiens adenosine kinase (ADK), transcript variant 3, mRNA.

1516


AICDA
NM_020661

Homo sapiens activation-induced cytidine deaminase (AICDA), mRNA.

1517


AK2
NM_001199199

Homo sapiens adenylate kinase 2 (AK2), transcript variant 3, mRNA.

1518


AK2
NM_013411

Homo sapiens adenylate kinase 2 (AK2), transcript variant 2, mRNA.

1519


AK2
NM_001625

Homo sapiens adenylate kinase 2 (AK2), transcript variant 1, mRNA.

1520


ALG12
NM_024105

Homo sapiens ALG12, alpha-1,6-mannosyltransferase (ALG12), mRNA.

1521


ALPL
NM_000478

Homo sapiens alkaline phosphatase, liver/bone/kidney (ALPL), transcript variant 1, mRNA.

1522


ALPL
NM_001127501

Homo sapiens alkaline phosphatase, liver/bone/kidney (ALPL), transcript variant 2, mRNA.

1523


ALPL
NM_001177520

Homo sapiens alkaline phosphatase, liver/bone/kidney (ALPL), transcript variant 3, mRNA.

1524


AP3B1
NM_001271769

Homo sapiens adaptor related protein complex 3 beta 1 subunit (AP3B1), transcript variant 2, mRNA.

1525


AP3B1
NM_003664

Homo sapiens adaptor related protein complex 3 beta 1 subunit (AP3B1), transcript variant 1, mRNA.

1526


AP3B2
NM_004644

Homo sapiens adaptor-related protein complex 3, beta 2 subunit (AP3B2), transcript variant 2, mRNA.

1527


AP3D1
NM_003938

Homo sapiens adaptor-related protein complex 3, delta 1 subunit (AP3D1), transcript variant 2, mRNA.

1528


AP3D1
NM_001261826

Homo sapiens adaptor-related protein complex 3, delta 1 subunit (AP3D1), transcript variant 3, mRNA.

1529


APOL1
NM_001136540

Homo sapiens apolipoprotein L1 (APOL1), transcript variant 3, mRNA.

1530


APOL1
NM_001136541

Homo sapiens apolipoprotein L1 (APOL1), transcript variant 4, mRNA.

1531


APOL1
NM_003661

Homo sapiens apolipoprotein L1 (APOL1), transcript variant 1, mRNA.

1532


APOL1
NM_145343

Homo sapiens apolipoprotein L1 (APOL1), transcript variant 2, mRNA.

1533


ASH1L
NM_018489

Homo sapiens ASH1 like histone lysine methyltransferase (ASH1L), mRNA.

1534


ATL2
NM_001135673

Homo sapiens atlastin GTPase 2 (ATL2), transcript variant 2, mRNA.

1535


ATL2
NM_022374

Homo sapiens atlastin GTPase 2 (ATL2), transcript variant 1, mRNA.

1536


ATL2
NR_024191

Homo sapiens atlastin GTPase 2 (ATL2), transcript variant 3, non-coding RNA.

1537


ATM
NM_000051

Homo sapiens ATM serine/threonine kinase (ATM), mRNA.

1538


ATR
NM_001184

Homo sapiens ATR serine/threonine kinase (ATR), mRNA.

1539


BACH2
NM_001170794

Homo sapiens BTB domain and CNC homolog 2 (BACH2), transcript variant 2, mRNA.

1540


BACH2
NM_021813

Homo sapiens BTB domain and CNC homolog 2 (BACH2), transcript variant 1, mRNA.

1541


BAG3
NM_004281

Homo sapiens BCL2 associated athanogene 3 (BAG3), mRNA.

1542


BCL10
NM_003921

Homo sapiens B-cell CLL/lymphoma 10 (BCL10), transcript variant 1, mRNA.

1543


BLM
NM_000057

Homo sapiens Bloom syndrome RecQ like helicase (BLM), transcript variant 1, mRNA.

1544


BLNK
NM_001114094

Homo sapiens B-cell linker (BLNK), transcript variant 2, mRNA.

1545


BLNK
NM_001258440

Homo sapiens B-cell linker (BLNK), transcript variant 3, mRNA.

1546


BLNK
NM_001258441

Homo sapiens B-cell linker (BLNK), transcript variant 4, mRNA.

1547


BLNK
NM_001258442

Homo sapiens B-cell linker (BLNK), transcript variant 5, mRNA.

1548


BLNK
NM_013314

Homo sapiens B-cell linker (BLNK), transcript variant 1, mRNA.

1549


BLNK
NR_047680

Homo sapiens B-cell linker (BLNK), transcript variant 6, non-coding RNA.

1550


BLNK
NR_047681

Homo sapiens B-cell linker (BLNK), transcript variant 7, non-coding RNA.

1551


BLNK
NR_047682

Homo sapiens B-cell linker (BLNK), transcript variant 8, non-coding RNA.

1552


BLNK
NR_047683

Homo sapiens B-cell linker (BLNK), transcript variant 9, non-coding RNA.

1553


BLOC1S6
NM_012388

Homo sapiens biogenesis of lysosomal organelles complex 1 subunit 6 (BLOC1S6), transcript variant 2, mRNA.

1554


BTK
NM_000061

Homo sapiens Bruton tyrosine kinase (BTK), transcript variant 1, mRNA.

1555


C11orf65
NM_152587

Homo sapiens chromosome 11 open reading frame 65 (C11orf65), mRNA.

1556


C1QA
NM_015991

Homo sapiens complement component 1, q subcomponent, A chain (C1QA), mRNA.

1557


C1QB
NM_000491

Homo sapiens complement component 1, q subcomponent, B chain (C1QB), mRNA.

1558


C1QC
NM_001114101

Homo sapiens complement component 1, q subcomponent, C chain (C1QC), transcript variant 1, mRNA.

1559


C1QC
NM_172369

Homo sapiens complement component 1, q subcomponent, C chain (C1QC), transcript variant 2, mRNA.

1560


C5AR1
NM_001736

Homo sapiens complement component 5a receptor 1 (C5AR1), mRNA.

1561


CARD11
NM_032415

Homo sapiens caspase recruitment domain family member 11 (CARD11), transcript variant 2, mRNA.

1562


CARD9
NM_052813

Homo sapiens caspase recruitment domain family, member 9 (CARD9), transcript variant 1, mRNA.

1563


CARD9
NM_052814

Homo sapiens caspase recruitment domain family, member 9 (CARD9), transcript variant 2, mRNA.

1564


CASP8
NM_001080124

Homo sapiens caspase 8 (CASP8), transcript variant F, mRNA.

1565


CASP8
NM_001228

Homo sapiens caspase 8 (CASP8), transcript variant A, mRNA.

1566


CASP8
NM_033355

Homo sapiens caspase 8 (CASP8), transcript variant B, mRNA.

1567


CASP8
NM_033358

Homo sapiens caspase 8 (CASP8), transcript variant E, mRNA.

1568


CASP8
NM_001080125

Homo sapiens caspase 8 (CASP8), transcript variant G, mRNA.

1569


CASP8
NM_033356

Homo sapiens caspase 8 (CASP8), transcript variant C, mRNA.

1570


CCL11
NM_002986

Homo sapiens C-C motif chemokine ligand 11 (CCL11), mRNA.

1571


CCL2
NM_002982

Homo sapiens C-C motif chemokine ligand 2 (CCL2), mRNA.

1572


CCL5
NM_002985

Homo sapiens C-C motif chemokine ligand 5 (CCL5), transcript variant 1, mRNA.

1573


CCR2
NM_001123041

Homo sapiens C-C motif chemokine receptor 2 (CCR2), transcript variant A, mRNA.

1574


CCR2
NM_001123396

Homo sapiens C-C motif chemokine receptor 2 (CCR2), transcript variant B, mRNA.

1575


CCR5
NM_000579

Homo sapiens C-C motif chemokine receptor 5 (gene/pseudogene) (CCR5), transcript variant A, mRNA.

1576


CCR5
NM_001100168

Homo sapiens C-C motif chemokine receptor 5 (gene/pseudogene) (CCR5), transcript variant B, mRNA.

1577


CD180
NM_005582

Homo sapiens CD180 molecule (CD180), mRNA.

1578


CD19
NM_001178098

Homo sapiens CD19 molecule (CD19), transcript variant 1, mRNA.

1579


CD19
NM_001770

Homo sapiens CD19 molecule (CD19), transcript variant 2, mRNA.

1580


CD209
NM_001144893

Homo sapiens CD209 molecule (CD209), transcript variant 5, mRNA.

1581


CD209
NM_001144894

Homo sapiens CD209 molecule (CD209), transcript variant 6, mRNA.

1582


CD209
NM_001144895

Homo sapiens CD209 molecule (CD209), transcript variant 7, mRNA.

1583


CD209
NM_001144896

Homo sapiens CD209 molecule (CD209), transcript variant 3, mRNA.

1584


CD209
NM_001144897

Homo sapiens CD209 molecule (CD209), transcript variant 4, mRNA.

1585


CD209
NM_001144899

Homo sapiens CD209 molecule (CD209), transcript variant 8, mRNA.

1586


CD209
NM_021155

Homo sapiens CD209 molecule (CD209), transcript variant 1, mRNA.

1587


CD209
NR_026692

Homo sapiens CD209 molecule (CD209), transcript variant 2, non-coding RNA.

1588


CD247
NM_000734

Homo sapiens CD247 molecule (CD247), transcript variant 2, mRNA.

1589


CD247
NM_198053

Homo sapiens CD247 molecule (CD247), transcript variant 1, mRNA.

1590


CD27
NM_001242

Homo sapiens CD27 molecule (CD27), mRNA.

1591


CD27-AS1
NR_015382

Homo sapiens CD27 antisense RNA 1 (CD27-AS1), long non-coding RNA.

1592


CD34
NM_001025109

Homo sapiens CD34 molecule (CD34), transcript variant 1, mRNA.

1593


CD34
NM_001773

Homo sapiens CD34 molecule (CD34), transcript variant 2, mRNA.

1594


CD3D
NM_000732

Homo sapiens CD3d molecule (CD3D), transcript variant 1, mRNA.

1595


CD3D
NM_001040651

Homo sapiens CD3d molecule (CD3D), transcript variant 2, mRNA.

1596


CD3E
NM_000733

Homo sapiens CD3e molecule (CD3E), mRNA.

1597


CD3G
NM_000073

Homo sapiens CD3g molecule (CD3G), mRNA.

1598


CD40
NM_001250

Homo sapiens CD40 molecule (CD40), transcript variant 1, mRNA.

1599


CD40
NM_152854

Homo sapiens CD40 molecule (CD40), transcript variant 2, mRNA.

1600


CD40LG
NM_000074

Homo sapiens CD40 ligand (CD40LG), mRNA.

1601


CD55
NM_000574

Homo sapiens CD55 molecule (Cromer blood group) (CD55), transcript variant 1, mRNA.

1602


CD55
NM_001114752

Homo sapiens CD55 molecule (Cromer blood group) (CD55), transcript variant 2, mRNA.

1603


CD59
NM_000611

Homo sapiens CD59 molecule (CD59), transcript variant 2, mRNA.

1604


CD59
NM_001127223

Homo sapiens CD59 molecule (CD59), transcript variant 5, mRNA.

1605


CD59
NM_001127225

Homo sapiens CD59 molecule (CD59), transcript variant 6, mRNA.

1606


CD59
NM_001127226

Homo sapiens CD59 molecule (CD59), transcript variant 7, mRNA.

1607


CD59
NM_001127227

Homo sapiens CD59 molecule (CD59), transcript variant 8, mRNA.

1608


CD59
NM_203329

Homo sapiens CD59 molecule (CD59), transcript variant 3, mRNA.

1609


CD59
NM_203330

Homo sapiens CD59 molecule (CD59), transcript variant 1, mRNA.

1610


CD59
NM_203331

Homo sapiens CD59 molecule (CD59), transcript variant 4, mRNA.

1611


CD79A
NM_001783

Homo sapiens CD79a molecule (CD79A), transcript variant 1, mRNA.

1612


CD79A
NM_021601

Homo sapiens CD79a molecule (CD79A), transcript variant 2, mRNA.

1613


CD79B
NM_000626

Homo sapiens CD79b molecule (CD79B), transcript variant 1, mRNA.

1614


CD79B
NM_001039933

Homo sapiens CD79b molecule (CD79B), transcript variant 3, mRNA.

1615


CD79B
NM_021602

Homo sapiens CD79b molecule (CD79B), transcript variant 2, mRNA.

1616


CD81
NM_004356

Homo sapiens CD81 molecule (CD81), transcript variant 1, mRNA.

1617


CD8A
NM_001145873

Homo sapiens CD8a molecule (CD8A), transcript variant 3, mRNA.

1618


CD8A
NM_001768

Homo sapiens CD8a molecule (CD8A), transcript variant 1, mRNA.

1619


CD8A
NM_171827

Homo sapiens CD8a molecule (CD8A), transcript variant 2, mRNA.

1620


CD8A
NR_027353

Homo sapiens CD8a molecule (CD8A), transcript variant 4, non-coding RNA.

1621


CDCA7
NM_031942

Homo sapiens cell division cycle associated 7 (CDCA7), transcript variant 1, mRNA.

1622


CDCA7
NM_145810

Homo sapiens cell division cycle associated 7 (CDCA7), transcript variant 2, mRNA.

1623


CEBPB
NM_005194

Homo sapiens CCAAT/enhancer binding protein beta (CEBPB), transcript variant 1, mRNA.

1624


CHD7
NM_017780

Homo sapiens chromodomain helicase DNA binding protein 7 (CHD7), transcript variant 1, mRNA.

1625


CHEK1
NM_001114121

Homo sapiens checkpoint kinase 1 (CHEK1), transcript variant 2, mRNA.

1626


CHEK1
NM_001114122

Homo sapiens checkpoint kinase 1 (CHEK1), transcript variant 1, mRNA.

1627


CHEK1
NM_001244846

Homo sapiens checkpoint kinase 1 (CHEK1), transcript variant 4, mRNA.

1628


CHEK1
NR_045204

Homo sapiens checkpoint kinase 1 (CHEK1), transcript variant 5, non-coding RNA.

1629


CHEK1
NR_045205

Homo sapiens checkpoint kinase 1 (CHEK1), transcript variant 6, non-coding RNA.

1630


CHEK1
NM_001274

Homo sapiens checkpoint kinase 1 (CHEK1), transcript variant 3, mRNA.

1631


CIITA
NM_000246

Homo sapiens class II major histocompatibility complex transactivator (CIITA), transcript variant 2, mRNA.

1632


CLCN7
NM_001114331

Homo sapiens chloride channel, voltage-sensitive 7 (CLCN7), transcript variant 2, mRNA.

1633


CLCN7
NM_001287

Homo sapiens chloride channel, voltage-sensitive 7 (CLCN7), transcript variant 1, mRNA.

1634


COG6
NM_001145079

Homo sapiens component of oligomeric golgi complex 6 (COG6), transcript variant 2, mRNA.

1635


COG6
NM_020751

Homo sapiens component of oligomeric golgi complex 6 (COG6), transcript variant 1, mRNA.

1636


COG6
NR_026745

Homo sapiens component of oligomeric golgi complex 6 (COG6), transcript variant 3, non-coding RNA.

1637


CORO1A
NM_001193333

Homo sapiens coronin 1A (CORO1A), transcript variant 1, mRNA.

1638


CORO1A
NM_007074

Homo sapiens coronin 1A (CORO1A), transcript variant 2, mRNA.

1639


CR2
NM_001006658

Homo sapiens complement component 3d receptor 2 (CR2), transcript variant 1, mRNA.

1640


CR2
NM_001877

Homo sapiens complement component 3d receptor 2 (CR2), transcript variant 2, mRNA.

1641


CRTC3
NM_001042574

Homo sapiens CREB regulated transcription coactivator 3 (CRTC3), transcript variant 2, mRNA.

1642


CRTC3
NM_022769

Homo sapiens CREB regulated transcription coactivator 3 (CRTC3), transcript variant 1, mRNA.

1643


CSF3R
NM_000760

Homo sapiens colony stimulating factor 3 receptor (granulocyte) (CSF3R), transcript variant 1, mRNA.

1644


CSF3R
NM_156039

Homo sapiens colony stimulating factor 3 receptor (granulocyte) (CSF3R), transcript variant 3, mRNA.

1645


CSF3R
NM_172313

Homo sapiens colony stimulating factor 3 receptor (granulocyte) (CSF3R), transcript variant 4, mRNA.

1646


CTLA4
NM_005214

Homo sapiens cytotoxic T-lymphocyte-associated protein 4 (CTLA4), transcript variant 1, mRNA.

1647


CTLA4
NM_001037631

Homo sapiens cytotoxic T-lymphocyte-associated protein 4 (CTLA4), transcript variant 2, mRNA.

1648


CTPS1
NM_001905

Homo sapiens CTP synthase 1 (CTPS1), transcript variant 1, mRNA.

1649


CTSC
NM_148170

Homo sapiens cathepsin C (CTSC), transcript variant 2, mRNA.

1650


CTSC
NM_001114173

Homo sapiens cathepsin C (CTSC), transcript variant 3, mRNA.

1651


CTSC
NM_001814

Homo sapiens cathepsin C (CTSC), transcript variant 1, mRNA.

1652


CX3CR1
NM_001171171

Homo sapiens C-X3-C motif chemokine receptor 1 (CX3CR1), transcript variant 2, mRNA.

1653


CX3CR1
NM_001171172

Homo sapiens C-X3-C motif chemokine receptor 1 (CX3CR1), transcript variant 3, mRNA.

1654


CX3CR1
NM_001171174

Homo sapiens C-X3-C motif chemokine receptor 1 (CX3CR1), transcript variant 1, mRNA.

1655


CX3CR1
NM_001337

Homo sapiens C-X3-C motif chemokine receptor 1 (CX3CR1), transcript variant 4, mRNA.

1656


CXCL12
NM_000609

Homo sapiens C-X-C motif chemokine ligand 12 (CXCL12), transcript variant 2, mRNA.

1657


CXCL12
NM_001033886

Homo sapiens C-X-C motif chemokine ligand 12 (CXCL12), transcript variant 3, mRNA.

1658


CXCL12
NM_001178134

Homo sapiens C-X-C motif chemokine ligand 12 (CXCL12), transcript variant 4, mRNA.

1659


CXCL12
NM_199168

Homo sapiens C-X-C motif chemokine ligand 12 (CXCL12), transcript variant 1, mRNA.

1660


CXCL9
NM_002416

Homo sapiens C-X-C motif chemokine ligand 9 (CXCL9), mRNA.

1661


CXCR1
NM_000634

Homo sapiens C-X-C motif chemokine receptor 1 (CXCR1), mRNA.

1662


CXCR4
NM_001008540

Homo sapiens C-X-C motif chemokine receptor 4 (CXCR4), transcript variant 1, mRNA.

1663


CXCR4
NM_003467

Homo sapiens C-X-C motif chemokine receptor 4 (CXCR4), transcript variant 2, mRNA.

1664


CXorf40A
NM_001171907

Homo sapiens chromosome X open reading frame 40A (CXorf40A), transcript variant 2, mRNA.

1665


CXorf40A
NM_001171908

Homo sapiens chromosome X open reading frame 40A (CXorf40A), transcript variant 3, mRNA.

1666


CXorf40A
NM_178124

Homo sapiens chromosome X open reading frame 40A (CXorf40A), transcript variant 1, mRNA.

1667


CXorf40A
NM_001171909

Homo sapiens chromosome X open reading frame 40A (CXorf40A), transcript variant 4, mRNA.

1668


CYBB
NM_000397

Homo sapiens cytochrome b-245, beta polypeptide (CYBB), mRNA.

1669


CYP2S1
NM_030622

Homo sapiens cytochrome P450 family 2 subfamily S member 1 (CYP2S1), mRNA.

1670


DCLRE1C
NM_001033855

Homo sapiens DNA cross-link repair 1C (DCLRE1C), transcript variant a, mRNA.

1671


DCLRE1C
NM_001033857

Homo sapiens DNA cross-link repair 1C (DCLRE1C), transcript variant d, mRNA.

1672


DCLRE1C
NM_001033858

Homo sapiens DNA cross-link repair 1C (DCLRE1C), transcript variant c, mRNA.

1673


DCLRE1C
NM_022487

Homo sapiens DNA cross-link repair 1C (DCLRE1C), transcript variant b, mRNA.

1674


DDX1
NM_004939

Homo sapiens DEAD/H-box helicase 1 (DDX1), mRNA.

1675


DDX58
NM_014314

Homo sapiens DEAD (Asp-Glu-Ala-Asp) box polypeptide 58 (DDX58), mRNA.

1676


DHX58
NM_024119

Homo sapiens DEXH (Asp-Glu-X-His) box polypeptide 58 (DHX58), mRNA.

1677


DKC1
NM_001142463

Homo sapiens dyskerin pseudouridine synthase 1 (DKC1), transcript variant 2, mRNA.

1678


DKC1
NM_001363

Homo sapiens dyskerin pseudouridine synthase 1 (DKC1), transcript variant 1, mRNA.

1679


DNMT3B
NM_001207055

Homo sapiens DNA methyltransferase 3 beta (DNMT3B), transcript variant 7, mRNA.

1680


DNMT3B
NM_001207056

Homo sapiens DNA methyltransferase 3 beta (DNMT3B), transcript variant 8, mRNA.

1681


DNMT3B
NM_006892

Homo sapiens DNA methyltransferase 3 beta (DNMT3B), transcript variant 1, mRNA.

1682


DNMT3B
NM_175848

Homo sapiens DNA methyltransferase 3 beta (DNMT3B), transcript variant 2, mRNA.

1683


DNMT3B
NM_175849

Homo sapiens DNA methyltransferase 3 beta (DNMT3B), transcript variant 3, mRNA.

1684


DNMT3B
NM_175850

Homo sapiens DNA methyltransferase 3 beta (DNMT3B), transcript variant 6, mRNA.

1685


DOCK2
NM_004946

Homo sapiens dedicator of cytokinesis 2 (DOCK2), mRNA.

1686


DOCK8
NM_203447

Homo sapiens dedicator of cytokinesis 8 (DOCK8), transcript variant 1, mRNA.

1687


DOCK8
NM_001190458

Homo sapiens dedicator of cytokinesis 8 (DOCK8), transcript variant 2, mRNA.

1688


DOCK8
NM_001193536

Homo sapiens dedicator of cytokinesis 8 (DOCK8), transcript variant 3, mRNA.

1689


DSC1
NM_004948

Homo sapiens desmocollin 1 (DSC1), transcript variant Dsc1b, mRNA.

1690


DSC1
NM_024421

Homo sapiens desmocollin 1 (DSC1), transcript variant Dsc1a, mRNA.

1691


EGR1
NM_001964

Homo sapiens early growth response 1 (EGR1), mRNA.

1692


ELANE
NM_001972

Homo sapiens elastase, neutrophil expressed (ELANE), mRNA.

1693


EPG5
NM_020964

Homo sapiens ectopic P-granules autophagy protein 5 homolog (EPG5), mRNA.

1694


ETF1
NM_004730

Homo sapiens eukaryotic translation termination factor 1 (ETF1), transcript variant 1, mRNA.

1695


F9
NM_000133

Homo sapiens coagulation factor IX (F9), transcript variant 1, mRNA.

1696


FAS
NM_000043

Homo sapiens Fas cell surface death receptor (FAS), transcript variant 1, mRNA.

1697


FAS
NM_152871

Homo sapiens Fas cell surface death receptor (FAS), transcript variant 2, mRNA.

1698


FAS
NM_152872

Homo sapiens Fas cell surface death receptor (FAS), transcript variant 3, mRNA.

1699


FAS
NR_028033

Homo sapiens Fas cell surface death receptor (FAS), transcript variant 4, non-coding RNA.

1700


FAS
NR_028034

Homo sapiens Fas cell surface death receptor (FAS), transcript variant 5, non-coding RNA.

1701


FAS
NR_028035

Homo sapiens Fas cell surface death receptor (FAS), transcript variant 6, non-coding RNA.

1702


FAS
NR_028036

Homo sapiens Fas cell surface death receptor (FAS), transcript variant 7, non-coding RNA.

1703


FASLG
NM_000639

Homo sapiens Fas ligand (TNF superfamily, member 6) (FASLG), mRNA.

1704


FCGR2A
NM_001136219

Homo sapiens Fc fragment of IgG receptor IIa (FCGR2A), transcript variant 1, mRNA.

1705


FCGR2A
NM_021642

Homo sapiens Fc fragment of IgG receptor IIa (FCGR2A), transcript variant 2, mRNA.

1706


FCGR3A
NM_000569

Homo sapiens Fc fragment of IgG receptor IIIa (FCGR3A), transcript variant 1, mRNA.

1707


FCGR3A
NM_001127592

Homo sapiens Fc fragment of IgG receptor IIIa (FCGR3A), transcript variant 2, mRNA.

1708


FCGR3A
NM_001127593

Homo sapiens Fc fragment of IgG receptor IIIa (FCGR3A), transcript variant 3, mRNA.

1709


FCGR3A
NM_001127595

Homo sapiens Fc fragment of IgG receptor IIIa (FCGR3A), transcript variant 4, mRNA.

1710


FCGR3A
NM_001127596

Homo sapiens Fc fragment of IgG receptor IIIa (FCGR3A), transcript variant 5, mRNA.

1711


FCN3
NM_003665

Homo sapiens ficolin 3 (FCN3), transcript variant 1, mRNA.

1712


FCN3
NM_173452

Homo sapiens ficolin 3 (FCN3), transcript variant 2, mRNA.

1713


FEZ1
NM_005103

Homo sapiens fasciculation and elongation protein zeta 1 (FEZ1), transcript variant 1, mRNA.

1714


FEZ1
NM_022549

Homo sapiens fasciculation and elongation protein zeta 1 (FEZ1), transcript variant 2, mRNA.

1715


FOS
NM_005252

Homo sapiens Fos proto-oncogene, AP-1 transcription factor subunit (FOS), mRNA.

1716


FOXH1
NM_003923

Homo sapiens forkhead box H1 (FOXH1), mRNA.

1717


FOXN1
NM_003593

Homo sapiens forkhead box N1 (FOXN1), mRNA.

1718


FOXP3
NM_001114377

Homo sapiens forkhead box P3 (FOXP3), transcript variant 2, mRNA.

1719


FOXP3
NM_014009

Homo sapiens forkhead box P3 (FOXP3), transcript variant 1, mRNA.

1720


FPR1
NM_001193306

Homo sapiens formyl peptide receptor 1 (FPR1), transcript variant 1, mRNA.

1721


FPR1
NM_002029

Homo sapiens formyl peptide receptor 1 (FPR1), transcript variant 2, mRNA.

1722


G6PC3
NM_138387

Homo sapiens glucose 6 phosphatase, catalytic, 3 (G6PC3), transcript variant 1, mRNA.

1723


G6PC3
NR_028582

Homo sapiens glucose 6 phosphatase, catalytic, 3 (G6PC3), transcript variant 2, non-coding RNA.

1724


G6PC3
NR_028581

Homo sapiens glucose 6 phosphatase, catalytic, 3 (G6PC3), transcript variant 3, non-coding RNA.

1725


GATA2
NM_001145661

Homo sapiens GATA binding protein 2 (GATA2), transcript variant 1, mRNA.

1726


GATA2
NM_001145662

Homo sapiens GATA binding protein 2 (GATA2), transcript variant 3, mRNA.

1727


GATA2
NM_032638

Homo sapiens GATA binding protein 2 (GATA2), transcript variant 2, mRNA.

1728


GFI1
NM_005263

Homo sapiens growth factor independent 1 transcription repressor (GFI1), transcript variant 1, mRNA.

1729


GFI1
NM_001127215

Homo sapiens growth factor independent 1 transcription repressor (GFI1), transcript variant 2, mRNA.

1730


GFI1
NM_001127216

Homo sapiens growth factor independent 1 transcription repressor (GFI1), transcript variant 3, mRNA.

1731


GOLGB1
NM_001256486

Homo sapiens golgin B1 (GOLGB1), transcript variant 1, mRNA.

1732


GOLGB1
NM_001256487

Homo sapiens golgin B1 (GOLGB1), transcript variant 3, mRNA.

1733


GOLGB1
NM_001256488

Homo sapiens golgin B1 (GOLGB1), transcript variant 4, mRNA.

1734


GOLGB1
NM_004487

Homo sapiens golgin B1 (GOLGB1), transcript variant 2, mRNA.

1735


GPRC5A
NM_003979

Homo sapiens G protein-coupled receptor class C group 5 member A (GPRC5A), mRNA.

1736


GRAP2
NM_004810

Homo sapiens GRB2-related adaptor protein 2 (GRAP2), mRNA.

1737


HAX1
NM_006118

Homo sapiens HCLS1 associated protein X-1 (HAX1), transcript variant 1, mRNA.

1738


HAX1
NM_001018837

Homo sapiens HCLS1 associated protein X-1 (HAX1), transcript variant 2, mRNA.

1739


HELLS
NM_018063

Homo sapiens helicase, lymphoid-specific (HELLS), transcript variant 1, mRNA.

1740


HIVEP1
NM_002114

Homo sapiens human immunodeficiency virus type I enhancer binding protein 1 (HIVEP1), mRNA.

1741


HIVEP2
NM_006734

Homo sapiens human immunodeficiency virus type I enhancer binding protein 2 (HIVEP2), mRNA.

1742


HIVEP3
NM_001127714

Homo sapiens human immunodeficiency virus type I enhancer binding protein 3 (HIVEP3), transcript variant 2, mRNA.

1743


HIVEP3
NM_024503

Homo sapiens human immunodeficiency virus type I enhancer binding protein 3 (HIVEP3), transcript variant 1, mRNA.

1744


HIVEP3
NR_038260

Homo sapiens human immunodeficiency virus type I enhancer binding protein 3 (HIVEP3), transcript variant 3, non-coding RNA.

1745


HIVEP3
NR_038261

Homo sapiens human immunodeficiency virus type I enhancer binding protein 3 (HIVEP3), transcript variant 4, non-coding RNA.

1746


HP
NM_001126102

Homo sapiens haptoglobin (HP), transcript variant 2, mRNA.

1747


HP
NM_005143

Homo sapiens haptoglobin (HP), transcript variant 1, mRNA.

1748


HPCAL1
NM_002149

Homo sapiens hippocalcin like 1 (HPCAL1), transcript variant 1, mRNA.

1749


HPCAL1
NM_134421

Homo sapiens hippocalcin like 1 (HPCAL1), transcript variant 2, mRNA.

1750


HPCAL1
NM_001258357

Homo sapiens hippocalcin like 1 (HPCAL1), transcript variant 3, mRNA.

1751


HPCAL1
NM_001258358

Homo sapiens hippocalcin like 1 (HPCAL1), transcript variant 4, mRNA.

1752


HPCAL1
NM_001258359

Homo sapiens hippocalcin like 1 (HPCAL1), transcript variant 5, mRNA.

1753


HTR2A
NM_000621

Homo sapiens 5-hydroxytryptamine receptor 2A (HTR2A), transcript variant 1, mRNA.

1754


HTR2A
NM_001165947

Homo sapiens 5-hydroxytryptamine (serotonin) receptor 2A, G protein-coupled (HTR2A), transcript variant 2, mRNA.

1755


ICOS
NM_012092

Homo sapiens inducible T-cell costimulator (ICOS), mRNA.

1756


IDI1
NM_004508

Homo sapiens isopentenyl-diphosphate delta isomerase 1 (IDI1), transcript variant 1, mRNA.

1757


IFIH1
NM_022168

Homo sapiens interferon induced with helicase C domain 1 (IFIH1), mRNA.

1758


IFNAR1
NM_000629

Homo sapiens interferon (alpha, beta and omega) receptor 1 (IFNAR1), mRNA.

1759


IFNAR2
NM_207584

Homo sapiens interferon (alpha, beta and omega) receptor 2 (IFNAR2), transcript variant 3, mRNA.

1760


IFNAR2
NM_207585

Homo sapiens interferon (alpha, beta and omega) receptor 2 (IFNAR2), transcript variant 1, mRNA.

1761


IFNAR2
NM_000874

Homo sapiens interferon (alpha, beta and omega) receptor 2 (IFNAR2), transcript variant 2, mRNA.

1762


IFNG
NM_000619

Homo sapiens interferon gamma (IFNG), mRNA.

1763


IFNGR1
NM_000416

Homo sapiens interferon gamma receptor 1 (IFNGR1), mRNA.

1764


IFNGR2
NM_005534

Homo sapiens interferon gamma receptor 2 (interferon gamma transducer 1) (IFNGR2), transcript variant 2, mRNA.

1765


IGLL1
NM_020070

Homo sapiens immunoglobulin lambda like polypeptide 1 (IGLL1), transcript variant 1, mRNA.

1766


IGLL1
NM_152855

Homo sapiens immunoglobulin lambda like polypeptide 1 (IGLL1), transcript variant 2, mRNA.

1767


IKBKB
NM_001190720

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKBKB), transcript variant 2, mRNA.

1768


IKBKB
NM_001242778

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKBKB), transcript variant 7, mRNA.

1769


IKBKB
NM_001556

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKBKB), transcript variant 1, mRNA.

1770


IKBKB
NR_033818

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKBKB), transcript variant 5, non-coding RNA.

1771


IKBKB
NR_033819

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKBKB), transcript variant 6, non-coding RNA.

1772


IKBKB
NR_040009

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase beta (IKBKB), transcript variant 8, non-coding RNA.

1773


IKBKG
NM_001099856

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase gamma (IKBKG), transcript variant 2, mRNA.

1774


IKBKG
NM_001099857

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase gamma (IKBKG), transcript variant 1, mRNA.

1775


IKBKG
NM_001145255

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase gamma (IKBKG), transcript variant 4, mRNA.

1776


IKBKG
NM_003639

Homo sapiens inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase gamma (IKBKG), transcript variant 3, mRNA.

1777


IKZF1
NM_001220765

Homo sapiens IKAROS family zinc finger 1 (IKZF1), transcript variant 2, mRNA.

1778


IKZF1
NM_001220767

Homo sapiens IKAROS family zinc finger 1 (IKZF1), transcript variant 4, mRNA.

1779


IKZF1
NM_001220768

Homo sapiens IKAROS family zinc finger 1 (IKZF1), transcript variant 5, mRNA.

1780


IKZF1
NM_001220770

Homo sapiens IKAROS family zinc finger 1 (IKZF1), transcript variant 7, mRNA.

1781


IKZF1
NM_001220771

Homo sapiens IKAROS family zinc finger 1 (IKZF1), transcript variant 8, mRNA.

1782


IKZF1
NM_006060

Homo sapiens IKAROS family zinc finger 1 (IKZF1), transcript variant 1, mRNA.

1783


IL10
NM_000572

Homo sapiens interleukin 10 (IL10), mRNA.

1784


IL10RA
NM_001558

Homo sapiens interleukin 10 receptor subunit alpha (IL10RA), transcript variant 1, mRNA.

1785


IL10RA
NR_026691

Homo sapiens interleukin 10 receptor subunit alpha (IL10RA), transcript variant 2, non-coding RNA.

1786


IL10RB
NM_000628

Homo sapiens interleukin 10 receptor subunit beta (IL10RB), mRNA.

1787


IL12B
NM_002187

Homo sapiens interleukin 12B (IL12B), mRNA.

1788


IL12RB1
NM_005535

Homo sapiens interleukin 12 receptor subunit beta 1 (IL12RB1), transcript variant 1, mRNA.

1789


IL12RB1
NM_153701

Homo sapiens interleukin 12 receptor subunit beta 1 (IL12RB1), transcript variant 2, mRNA.

1790


IL17F
NM_052872

Homo sapiens interleukin 17F (IL17F), mRNA.

1791


IL17RA
NM_014339

Homo sapiens interleukin 17 receptor A (IL17RA), transcript variant 1, mRNA.

1792


IL1B
NM_000576

Homo sapiens interleukin 1, beta (IL1B), mRNA.

1793


IL21
NM_001207006

Homo sapiens interleukin 21 (IL21), transcript variant 2, mRNA.

1794


IL21
NM_021803

Homo sapiens interleukin 21 (IL21), transcript variant 1, mRNA.

1795


IL21R
NM_181078

Homo sapiens interleukin 21 receptor (IL21R), transcript variant 2, mRNA.

1796


IL21R
NM_181079

Homo sapiens interleukin 21 receptor (IL21R), transcript variant 3, mRNA.

1797


IL21R
NM_021798

Homo sapiens interleukin 21 receptor (IL21R), transcript variant 1, mRNA.

1798


IL2RA
NM_000417

Homo sapiens interleukin 2 receptor, alpha (IL2RA), transcript variant 1, mRNA.

1799


IL2RG
NM_000206

Homo sapiens interleukin 2 receptor subunit gamma (IL2RG), mRNA.

1800


IL4R
NM_000418

Homo sapiens interleukin 4 receptor (IL4R), transcript variant 1, mRNA.

1801


IL4R
NM_001257406

Homo sapiens interleukin 4 receptor (IL4R), transcript variant 3, mRNA.

1802


IL4R
NM_001257407

Homo sapiens interleukin 4 receptor (IL4R), transcript variant 4, mRNA.

1803


IL4R
NM_001257997

Homo sapiens interleukin 4 receptor (IL4R), transcript variant 5, mRNA.

1804


IL7
NM_000880

Homo sapiens interleukin 7 (IL7), transcript variant 1, mRNA.

1805


IL7
NM_001199886

Homo sapiens interleukin 7 (IL7), transcript variant 2, mRNA.

1806


IL7
NM_001199887

Homo sapiens interleukin 7 (IL7), transcript variant 3, mRNA.

1807


IL7
NM_001199888

Homo sapiens interleukin 7 (IL7), transcript variant 4, mRNA.

1808


IL7R
NM_002185

Homo sapiens interleukin 7 receptor (IL7R), transcript variant 1, mRNA.

1809


IRAK4
NM_001114182

Homo sapiens interleukin 1 receptor associated kinase 4 (IRAK4), transcript variant 1, mRNA.

1810


IRAK4
NM_001145256

Homo sapiens interleukin 1 receptor associated kinase 4 (IRAK4), transcript variant 3, mRNA.

1811


IRAK4
NM_001145257

Homo sapiens interleukin 1 receptor associated kinase 4 (IRAK4), transcript variant 4, mRNA.

1812


IRAK4
NM_001145258

Homo sapiens interleukin 1 receptor associated kinase 4 (IRAK4), transcript variant 5, mRNA.

1813


IRAK4
NM_016123

Homo sapiens interleukin 1 receptor associated kinase 4 (IRAK4), transcript variant 2, mRNA.

1814


IRF3
NM_001197122

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 2, mRNA.

1815


IRF3
NM_001197123

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 3, mRNA.

1816


IRF3
NM_001197124

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 4, mRNA.

1817


IRF3
NM_001197125

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 5, mRNA.

1818


IRF3
NM_001197126

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 6, mRNA.

1819


IRF3
NM_001197127

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 7, mRNA.

1820


IRF3
NM_001197128

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 8, mRNA.

1821


IRF3
NM_001571

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 1, mRNA.

1822


IRF3
NR_045568

Homo sapiens interferon regulatory factor 3 (IRF3), transcript variant 9, non-coding RNA.

1823


IRF7
NM_001572

Homo sapiens interferon regulatory factor 7 (IRF7), transcript variant a, mRNA.

1824


IRF7
NM_004029

Homo sapiens interferon regulatory factor 7 (IRF7), transcript variant b, mRNA.

1825


IRF7
NM_004031

Homo sapiens interferon regulatory factor 7 (IRF7), transcript variant d, mRNA.

1826


IRF8
NM_002163

Homo sapiens interferon regulatory factor 8 (IRF8), mRNA.

1827


IRGM
NM_001145805

Homo sapiens immunity related GTPase M (IRGM), mRNA.

1828


ISG15
NM_005101

Homo sapiens ISG15 ubiquitin-like modifier (ISG15), mRNA.

1829


ITK
NM_005546

Homo sapiens IL2 inducible T-cell kinase (ITK), mRNA.

1830


ITSN2
NM_006277

Homo sapiens intersectin 2 (ITSN2), transcript variant 1, mRNA.

1831


ITSN2
NM_019595

Homo sapiens intersectin 2 (ITSN2), transcript variant 3, mRNA.

1832


ITSN2
NM_147152

Homo sapiens intersectin 2 (ITSN2), transcript variant 2, mRNA.

1833


JAGN1
NM_032492

Homo sapiens jagunal homolog 1 (Drosophila) (JAGN1), mRNA.

1834


JAK3
NM_000215

Homo sapiens Janus kinase 3 (JAK3), mRNA.

1835


JMY
NM_152405

Homo sapiens junction mediating and regulatory protein, p53 cofactor (JMY), mRNA.

1836


JUN
NM_002228

Homo sapiens Jun proto-oncogene, AP-1 transcription factor subunit (JUN), mRNA.

1837


KITLG
NM_000899

Homo sapiens KIT ligand (KITLG), transcript variant b, mRNA.

1838


KITLG
NM_003994

Homo sapiens KIT ligand (KITLG), transcript variant a, mRNA.

1839


LAMTOR2
NM_001145264

Homo sapiens late endosomal/lysosomal adaptor, MAPK and MTOR activator 2 (LAMTOR2), transcript variant 2, mRNA.

1840


LAMTOR2
NM_014017

Homo sapiens late endosomal/lysosomal adaptor, MAPK and MTOR activator 2 (LAMTOR2), transcript variant 1, mRNA.

1841


LCK
NM_005356

Homo sapiens LCK proto-oncogene, Src family tyrosine kinase (LCK), transcript variant 2, mRNA.

1842


LCK
NM_001042771

Homo sapiens LCK proto-oncogene, Src family tyrosine kinase (LCK), transcript variant 1, mRNA.

1843


LCP2
NM_005565

Homo sapiens lymphocyte cytosolic protein 2 (SH2 domain containing leukocyte protein of 76 kDa) (LCP2), mRNA.

1844


LIG1
NM_000234

Homo sapiens DNA ligase 1 (LIG1), transcript variant 1, mRNA.

1845


LIG4
NM_001098268

Homo sapiens DNA ligase 4 (LIG4), transcript variant 3, mRNA.

1846


LIG4
NM_002312

Homo sapiens DNA ligase 4 (LIG4), transcript variant 1, mRNA.

1847


LIG4
NM_206937

Homo sapiens DNA ligase 4 (LIG4), transcript variant 2, mRNA.

1848


LRBA
NM_001199282

Homo sapiens LPS responsive beige-like anchor protein (LRBA), transcript variant 1, mRNA.

1849


LRBA
NM_006726

Homo sapiens LPS responsive beige-like anchor protein (LRBA), transcript variant 2, mRNA.

1850


LYST
NM_000081

Homo sapiens lysosomal trafficking regulator (LYST), transcript variant 1, mRNA.

1851


MAGEA9
NM_005365

Homo sapiens MAGE family member A9 (MAGEA9), mRNA.

1852


MAGEA9B
NM_001080790

Homo sapiens MAGE family member A9B (MAGEA9B), mRNA.

1853


MAGT1
NM_032121

Homo sapiens magnesium transporter 1 (MAGT1), mRNA.

1854


MALT1
NM_006785

Homo sapiens MALT1 paracaspase (MALT1), transcript variant 1, mRNA.

1855


MALT1
NM_173844

Homo sapiens MALT1 paracaspase (MALT1), transcript variant 2, mRNA.

1856


MAP3K2
NM_006609

Homo sapiens mitogen-activated protein kinase kinase kinase 2 (MAP3K2), mRNA.

1857


MAPK1
NM_002745

Homo sapiens mitogen-activated protein kinase 1 (MAPK1), transcript variant 1, mRNA.

1858


MAPK1
NM_138957

Homo sapiens mitogen-activated protein kinase 1 (MAPK1), transcript variant 2, mRNA.

1859


MAPK3
NM_001040056

Homo sapiens mitogen-activated protein kinase 3 (MAPK3), transcript variant 2, mRNA.

1860


MAPK3
NM_001109891

Homo sapiens mitogen-activated protein kinase 3 (MAPK3), transcript variant 3, mRNA.

1861


MAPK3
NM_002746

Homo sapiens mitogen-activated protein kinase 3 (MAPK3), transcript variant 1, mRNA.

1862


MAVS
NM_020746

Homo sapiens mitochondrial antiviral signaling protein (MAVS), transcript variant 1, mRNA.

1863


MAVS
NM_001206491

Homo sapiens mitochondrial antiviral signaling protein (MAVS), transcript variant 3, mRNA.

1864


MAVS
NR_037921

Homo sapiens mitochondrial antiviral signaling protein (MAVS), transcript variant 2, non-coding RNA.

1865


MECP2
NM_004992

Homo sapiens methyl-CpG binding protein 2 (MECP2), transcript variant 1, mRNA.

1866


MECP2
NM_001110792

Homo sapiens methyl-CpG binding protein 2 (MECP2), transcript variant 2, mRNA.

1867


MEX3C
NM_016626

Homo sapiens mex-3 RNA binding family member C (MEX3C), mRNA.

1868


MRE11A
NM_005590

Homo sapiens MRE11 homolog A, double strand break repair nuclease (MRE11A), transcript variant 2, mRNA.

1869


MRE11A
NM_005591

Homo sapiens MRE11 homolog A, double strand break repair nuclease (MRE11A), transcript variant 1, mRNA.

1870


MS4A1
NM_021950

Homo sapiens membrane spanning 4-domains A1 (MS4A1), transcript variant 3, mRNA.

1871


MS4A1
NM_152866

Homo sapiens membrane spanning 4-domains A1 (MS4A1), transcript variant 1, mRNA.

1872


MSN
NM_002444

Homo sapiens moesin (MSN), mRNA.

1873


MYD88
NM_001172566

Homo sapiens myeloid differentiation primary response 88 (MYD88), transcript variant 5, mRNA.

1874


MYD88
NM_001172567

Homo sapiens myeloid differentiation primary response 88 (MYD88), transcript variant 1, mRNA.

1875


MYD88
NM_001172568

Homo sapiens myeloid differentiation primary response 88 (MYD88), transcript variant 3, mRNA.

1876


MYD88
NM_001172569

Homo sapiens myeloid differentiation primary response 88 (MYD88), transcript variant 4, mRNA.

1877


MYD88
NM_002468

Homo sapiens myeloid differentiation primary response 88 (MYD88), transcript variant 2, mRNA.

1878


NBN
NM_002485

Homo sapiens nibrin (NBN), mRNA.

1879


NFIC
NM_001245005

Homo sapiens nuclear factor I C (NFIC), transcript variant 4, mRNA.

1880


NFIC
NM_205843

Homo sapiens nuclear factor I C (NFIC), transcript variant 2, mRNA.

1881


NFIC
NM_001245002

Homo sapiens nuclear factor I C (NFIC), transcript variant 1, mRNA.

1882


NFIC
NM_001245004

Homo sapiens nuclear factor I C (NFIC), transcript variant 3, mRNA.

1883


NFIC
NM_005597

Homo sapiens nuclear factor I C (NFIC), transcript variant 5, mRNA.

1884


NFKB1
NM_003998

Homo sapiens nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1), transcript variant 1, mRNA.

1885


NFKB1
NM_001165412

Homo sapiens nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1), transcript variant 2, mRNA.

1886


NFKB2
NM_001077494

Homo sapiens nuclear factor of kappa light polypeptide gene enhancer in B-cells 2 (NFKB2), transcript variant 1, mRNA.

1887


NFKB2
NM_002502

Homo sapiens nuclear factor of kappa light polypeptide gene enhancer in B-cells 2 (NFKB2), transcript variant 2, mRNA.

1888


NFKB2
NM_001261403

Homo sapiens nuclear factor of kappa light polypeptide gene enhancer in B-cells 2 (NFKB2), transcript variant 4, mRNA.

1889


NFKBIA
NM_020529

Homo sapiens NFKB inhibitor alpha (NFKBIA), mRNA.

1890


NHEJ1
NM_024782

Homo sapiens non-homologous end joining factor 1 (NHEJ1), mRNA.

1891


NLRP3
NM_183395

Homo sapiens NLR family, pyrin domain containing 3 (NLRP3), transcript variant 2, mRNA.

1892


NLRP3
NM_004895

Homo sapiens NLR family, pyrin domain containing 3 (NLRP3), transcript variant 1, mRNA.

1893


NLRP3
NM_001127462

Homo sapiens NLR family, pyrin domain containing 3 (NLRP3), transcript variant 5, mRNA.

1894


NLRP3
NM_001127461

Homo sapiens NLR family, pyrin domain containing 3 (NLRP3), transcript variant 4, mRNA.

1895


NLRP3
NM_001079821

Homo sapiens NLR family, pyrin domain containing 3 (NLRP3), transcript variant 3, mRNA.

1896


NLRP3
NM_001243133

Homo sapiens NLR family, pyrin domain containing 3 (NLRP3), transcript variant 6, mRNA.

1897


NOD2
NM_022162

Homo sapiens nucleotide-binding oligomerization domain containing 2 (NOD2), mRNA.

1898


ORAI1
NM_032790

Homo sapiens ORAI calcium release-activated calcium modulator 1 (ORAI1), mRNA.

1899


OSTM1
NM_014028

Homo sapiens osteopetrosis associated transmembrane protein 1 (OSTM1), mRNA.

1900


PGM3
NM_001199917

Homo sapiens phosphoglucomutase 3 (PGM3), transcript variant 1, mRNA.

1901


PGM3
NM_001199918

Homo sapiens phosphoglucomutase 3 (PGM3), transcript variant 3, mRNA.

1902


PGM3
NM_015599

Homo sapiens phosphoglucomutase 3 (PGM3), transcript variant 2, mRNA.

1903


PGM3
NM_001199919

Homo sapiens phosphoglucomutase 3 (PGM3), transcript variant 4, mRNA.

1904


PIAS1
NM_016166

Homo sapiens protein inhibitor of activated STAT1 (PIAS1), transcript variant 2, mRNA.

1905


PIK3R1
NM_181523

Homo sapiens phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), transcript variant 1, mRNA.

1906


PIK3R1
NM_181524

Homo sapiens phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), transcript variant 3, mRNA.

1907


PIK3R1
NM_181504

Homo sapiens phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), transcript variant 2, mRNA.

1908


PIK3R1
NM_001242466

Homo sapiens phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), transcript variant 4, mRNA.

1909


PLCG2
NM_002661

Homo sapiens phospholipase C gamma 2 (PLCG2), mRNA.

1910


PMS2
NM_000535

Homo sapiens PMS1 homolog 2, mismatch repair system component (PMS2), transcript variant 1, mRNA.

1911


PNP
NM_000270

Homo sapiens purine nucleoside phosphorylase (PNP), mRNA.

1912


POLA1
NM_016937

Homo sapiens polymerase (DNA directed), alpha 1, catalytic subunit (POLA1), mRNA.

1913


POLE
NM_006231

Homo sapiens DNA polymerase epsilon, catalytic subunit (POLE), mRNA.

1914


PRF1
NM_001083116

Homo sapiens perform 1 (PRF1), transcript variant 2, mRNA.

1915


PRF1
NM_005041

Homo sapiens perform 1 (PRF1), transcript variant 1, mRNA.

1916


PRKCD
NM_006254

Homo sapiens protein kinase C delta (PRKCD), transcript variant 1, mRNA.

1917


PRKCD
NM_212539

Homo sapiens protein kinase C delta (PRKCD), transcript variant 2, mRNA.

1918


PRKDC
NM_001081640

Homo sapiens protein kinase, DNA-activated, catalytic polypeptide (PRKDC), transcript variant 2, mRNA.

1919


PRKDC
NM_006904

Homo sapiens protein kinase, DNA-activated, catalytic polypeptide (PRKDC), transcript variant 1, mRNA.

1920


PROC
NM_000312

Homo sapiens protein C, inactivator of coagulation factors Va and VIIIa (PROC), mRNA.

1921


PSMB8
NM_004159

Homo sapiens proteasome (prosome, macropain) subunit, beta type, 8 (PSMB8), transcript variant 1, mRNA.

1922


PSMB8
NM_148919

Homo sapiens proteasome (prosome, macropain) subunit, beta type, 8 (PSMB8), transcript variant 2, mRNA.

1923


PTEN
NM_000314

Homo sapiens phosphatase and tensin homolog (PTEN), transcript variant 1, mRNA.

1924


PTPRC
NM_001267798

Homo sapiens protein tyrosine phosphatase, receptor type C (PTPRC), transcript variant 5, mRNA.

1925


PTPRC
NM_002838

Homo sapiens protein tyrosine phosphatase, receptor type C (PTPRC), transcript variant 1, mRNA.

1926


PTPRC
NM_080921

Homo sapiens protein tyrosine phosphatase, receptor type C (PTPRC), transcript variant 2, mRNA.

1927


PTPRC
NR_052021

Homo sapiens protein tyrosine phosphatase, receptor type C (PTPRC), transcript variant 4, non-coding RNA.

1928


PURA
NM_005859

Homo sapiens purine rich element binding protein A (PURA), mRNA.

1929


RAB27A
NM_183235

Homo sapiens RAB27A, member RAS oncogene family (RAB27A), transcript variant 3, mRNA.

1930


RAB27A
NM_183236

Homo sapiens RAB27A, member RAS oncogene family (RAB27A), transcript variant 4, mRNA.

1931


RAB27A
NM_004580

Homo sapiens RAB27A, member RAS oncogene family (RAB27A), transcript variant 1, mRNA.

1932


RAB27A
NM_183234

Homo sapiens RAB27A, member RAS oncogene family (RAB27A), transcript variant 2, mRNA.

1933


RAB7A
NM_004637

Homo sapiens RAB7A, member RAS oncogene family (RAB7A), mRNA.

1934


RABGEF1
NM_014504

Homo sapiens RAB guanine nucleotide exchange factor (GEF) 1 (RABGEF1), transcript variant 4, mRNA.

1935


RAC2
NM_002872

Homo sapiens ras-related C3 botulinum toxin substrate 2 (rho family, small GTP binding protein Rac2) (RAC2), mRNA.

1936


RAD51
NM_001164270

Homo sapiens RAD51 recombinase (RAD51), transcript variant 3, mRNA.

1937


RAD51
NM_002875

Homo sapiens RAD51 recombinase (RAD51), transcript variant 1, mRNA.

1938


RAD51
NM_133487

Homo sapiens RAD51 recombinase (RAD51), transcript variant 2, mRNA.

1939


RAD51
NM_001164269

Homo sapiens RAD51 recombinase (RAD51), transcript variant 4, mRNA.

1940


RAG1
NM_000448

Homo sapiens recombination activating gene 1 (RAG1), mRNA.

1941


RAG2
NM_000536

Homo sapiens recombination activating gene 2 (RAG2), transcript variant 1, mRNA.

1942


RAG2
NM_001243785

Homo sapiens recombination activating gene 2 (RAG2), transcript variant 3, mRNA.

1943


RAG2
NM_001243786

Homo sapiens recombination activating gene 2 (RAG2), transcript variant 4, mRNA.

1944


RBCK1
NM_006462

Homo sapiens RANBP2-type and C3HC4-type zinc finger containing 1 (RBCK1), transcript variant 1, mRNA.

1945


RBCK1
NM_031229

Homo sapiens RANBP2-type and C3HC4-type zinc finger containing 1 (RBCK1), transcript variant 2, mRNA.

1946


RFX5
NM_000449

Homo sapiens regulatory factor X5 (RFX5), transcript variant 1, mRNA.

1947


RFX5
NM_001025603

Homo sapiens regulatory factor X5 (RFX5), transcript variant 2, mRNA.

1948


RFXANK
NM_003721

Homo sapiens regulatory factor X associated ankyrin containing protein (RFXANK), transcript variant 1, mRNA.

1949


RFXANK
NM_134440

Homo sapiens regulatory factor X associated ankyrin containing protein (RFXANK), transcript variant 2, mRNA.

1950


RFXAP
NM_000538

Homo sapiens regulatory factor X associated protein (RFXAP), mRNA.

1951


RIPK1
NM_003804

Homo sapiens receptor (TNFRSF)-interacting serine-threonine kinase 1 (RIPK1), mRNA.

1952


RIPK3
NM_006871

Homo sapiens receptor-interacting serine-threonine kinase 3 (RIPK3), mRNA.

1953


RMRP
NR_003051

Homo sapiens RNA component of mitochondrial RNA processing endoribonuclease (RMRP), RNase MRP RNA.

1954


RNASEH2A
NM_006397

Homo sapiens ribonuclease H2, subunit A (RNASEH2A), mRNA.

1955


RNASEH2B
NM_001142279

Homo sapiens ribonuclease H2, subunit B (RNASEH2B), transcript variant 2, mRNA.

1956


RNASEH2B
NM_024570

Homo sapiens ribonuclease H2, subunit B (RNASEH2B), transcript variant 1, mRNA.

1957


RNASEH2C
NM_032193

Homo sapiens ribonuclease H2, subunit C (RNASEH2C), mRNA.

1958


RNASEL
NM_021133

Homo sapiens ribonuclease L (2′,5′-oligoisoadenylate synthetase-dependent) (RNASEL), mRNA.

1959


RNF168
NM_152617

Homo sapiens ring finger protein 168 (RNF168), mRNA.

1960


RNF31
NM_017999

Homo sapiens ring finger protein 31 (RNF31), mRNA.

1961


RNU4ATAC
NR_023343

Homo sapiens RNA, U4atac small nuclear (U12-dependent splicing) (RNU4ATAC), small nuclear RNA.

1962


RTEL1
NM_016434

Homo sapiens regulator of telomere elongation helicase 1 (RTEL1), transcript variant 1, mRNA.

1963


RTEL1
NM_032957

Homo sapiens regulator of telomere elongation helicase 1 (RTEL1), transcript variant 2, mRNA.

1964


RTEL1-
NR_037882

Homo sapiens RTEL1-TNFRSF6B readthrough (NMD candidate) (RTEL1-TNFRSF6B), long non-coding RNA.

1965


TNFRSF6B





SALL2
NM_005407

Homo sapiens spalt like transcription factor 2 (SALL2), transcript variant 1, mRNA.

1966


SAMHD1
NM_015474

Homo sapiens SAM domain and HD domain 1 (SAMHD1), mRNA.

1967


SBDS
NM_016038

Homo sapiens Shwachman-Bodian-Diamond syndrome (SBDS), mRNA.

1968


SH2D1A
NM_001114937

Homo sapiens SH2 domain containing 1A (SH2D1A), transcript variant 2, mRNA.

1969


SH2D1A
NM_002351

Homo sapiens SH2 domain containing 1A (SH2D1A), transcript variant 1, mRNA.

1970


SHARPIN
NM_030974

Homo sapiens SHANK-associated RH domain interactor (SHARPIN), transcript variant 1, mRNA.

1971


SHARPIN
NR_038270

Homo sapiens SHANK-associated RH domain interactor (SHARPIN), transcript variant 2, non-coding RNA.

1972


SKIV2L
NM_006929

Homo sapiens superkiller viralicidic activity 2-like (S. cerevisiae) (SKIV2L), mRNA.

1973


SLC37A4
NM_001164277

Homo sapiens solute carrier family 37 (glucose-6-phosphate transporter), member 4 (SLC37A4), transcript variant 1, mRNA.

1974


SLC37A4
NM_001164278

Homo sapiens solute carrier family 37 (glucose-6-phosphate transporter), member 4 (SLC37A4), transcript variant 2, mRNA.

1975


SLC37A4
NM_001164279

Homo sapiens solute carrier family 37 (glucose-6-phosphate transporter), member 4 (SLC37A4), transcript variant 3, mRNA.

1976


SLC37A4
NM_001467

Homo sapiens solute carrier family 37 (glucose-6-phosphate transporter), member 4 (SLC37A4), transcript variant 4, mRNA.

1977


SLC37A4
NM_001164280

Homo sapiens solute carrier family 37 (glucose-6-phosphate transporter), member 4 (SLC37A4), transcript variant 5, mRNA.

1978


SLC46A1
NM_001242366

Homo sapiens solute carrier family 46 member 1 (SLC46A1), transcript variant 2, mRNA.

1979


SLC46A1
NM_080669

Homo sapiens solute carrier family 46 member 1 (SLC46A1), transcript variant 1, mRNA.

1980


SLC8A1
NM_001112800

Homo sapiens solute carrier family 8 member A1 (SLC8A1), transcript variant B, mRNA.

1981


SLC8A1
NM_001112801

Homo sapiens solute carrier family 8 member A1 (SLC8A1), transcript variant C, mRNA.

1982


SLC8A1
NM_001112802

Homo sapiens solute carrier family 8 member A1 (SLC8A1), transcript variant D, mRNA.

1983


SLC8A1
NM_001252624

Homo sapiens solute carrier family 8 member A1 (SLC8A1), transcript variant E, mRNA.

1984


SLC8A1
NM_021097

Homo sapiens solute carrier family 8 member A1 (SLC8A1), transcript variant A, mRNA.

1985


SMAD2
NM_001003652

Homo sapiens SMAD family member 2 (SMAD2), transcript variant 2, mRNA.

1986


SMAD2
NM_001135937

Homo sapiens SMAD family member 2 (SMAD2), transcript variant 3, mRNA.

1987


SMAD2
NM_005901

Homo sapiens SMAD family member 2 (SMAD2), transcript variant 1, mRNA.

1988


SMAD3
NM_005902

Homo sapiens SMAD family member 3 (SMAD3), transcript variant 1, mRNA.

1989


SMAD3
NM_001145102

Homo sapiens SMAD family member 3 (SMAD3), transcript variant 2, mRNA.

1990


SMAD3
NM_001145103

Homo sapiens SMAD family member 3 (SMAD3), transcript variant 3, mRNA.

1991


SMAD3
NM_001145104

Homo sapiens SMAD family member 3 (SMAD3), transcript variant 4, mRNA.

1992


SMAD4
NM_005359

Homo sapiens SMAD family member 4 (SMAD4), mRNA.

1993


SNAP29
NM_004782

Homo sapiens synaptosomal-associated protein, 29 kDa (SNAP29), mRNA.

1994


SNAR-A1
NR_004435

Homo sapiens small ILF3/NF90-associated RNA A1 (SNAR-A1), small nuclear RNA.

1995


SNAR-A10
NR_024229

Homo sapiens small ILF3/NF90-associated RNA A10 (SNAR-A10), small nuclear RNA.

1996


SNAR-A11
NR_024225

Homo sapiens small ILF3/NF90-associated RNA A11 (SNAR-A11), small nuclear RNA.

1997


SNAR-A12
NR_004437

Homo sapiens small ILF3/NF90-associated RNA A12 (SNAR-A12), small nuclear RNA.

1998


SNAR-A13
NR_024216

Homo sapiens small ILF3/NF90-associated RNA A13 (SNAR-A13), small nuclear RNA.

1999


SNAR-A14
NR_024242

Homo sapiens small ILF3/NF90-associated RNA A14 (SNAR-A14), small nuclear RNA.

2000


SNAR-A2
NR_004436

Homo sapiens small ILF3/NF90-associated RNA A2 (SNAR-A2), small nuclear RNA.

2001


SNAR-A3
NR_024214

Homo sapiens small ILF3/NF90-associated RNA A3 (SNAR-A3), small nuclear RNA.

2002


SNAR-A4
NR_024215

Homo sapiens small ILF3/NF90-associated RNA A4 (SNAR-A4), small nuclear RNA.

2003


SNAR-A5
NR_024223

Homo sapiens small ILF3/NF90-associated RNA A5 (SNAR-A5), small nuclear RNA.

2004


SNAR-A6
NR_024227

Homo sapiens small ILF3/NF90-associated RNA A6 (SNAR-A6), small nuclear RNA.

2005


SNAR-A7
NR_024224

Homo sapiens small ILF3/NF90-associated RNA A7 (SNAR-A7), small nuclear RNA.

2006


SNAR-A8
NR_024228

Homo sapiens small ILF3/NF90-associated RNA A8 (SNAR-A8), small nuclear RNA.

2007


SNAR-A9
NR_024226

Homo sapiens small ILF3/NF90-associated RNA A9 (SNAR-A9), small nuclear RNA.

2008


SNAR-B1
NR_024231

Homo sapiens small ILF3/NF90-associated RNA B1 (SNAR-B1), small nuclear RNA.

2009


SNAR-B2
NR_024230

Homo sapiens small ILF3/NF90-associated RNA B2 (SNAR-B2), small nuclear RNA.

2010


SNAR-C1
NR_024220

Homo sapiens small ILF3/NF90-associated RNA C1 (SNAR-C1), small nuclear RNA.

2011


SNAR-C2
NR_024217

Homo sapiens small ILF3/NF90-associated RNA C2 (SNAR-C2), small nuclear RNA.

2012


SNAR-C3
NR_024221

Homo sapiens small ILF3/NF90-associated RNA C3 (SNAR-C3), small nuclear RNA.

2013


SNAR-C4
NR_024218

Homo sapiens small ILF3/NF90-associated RNA C4 (SNAR-C4), small nuclear RNA.

2014


SNAR-C5
NR_024219

Homo sapiens small ILF3/NF90-associated RNA C5 (SNAR-C5), small nuclear RNA.

2015


SNAR-D
NR_024243

Homo sapiens small ILF3/NF90-associated RNA D (SNAR-D), small nuclear RNA.

2016


SNAR-E
NR_024258

Homo sapiens small ILF3/NF90-associated RNA E (SNAR-E), small nuclear RNA.

2017


SNAR-F
NR_004384

Homo sapiens small ILF3/NF90-associated RNA F (SNAR-F), small nuclear RNA.

2018


SNAR-G1
NR_004383

Homo sapiens small ILF3/NF90-associated RNA G1 (SNAR-G1), small nuclear RNA.

2019


SNAR-G2
NR_024244

Homo sapiens small ILF3/NF90-associated RNA G2 (SNAR-G2), small nuclear RNA.

2020


SNAR-H
NR_024342

Homo sapiens small ILF3/NF90-associated RNA H (SNAR-H), small nuclear RNA.

2021


SNAR-I
NR_024343

Homo sapiens small ILF3/NF90-associated RNA I (SNAR-I), small nuclear RNA.

2022


SNCA
NM_000345

Homo sapiens synuclein, alpha (non A4 component of amyloid precursor) (SNCA), transcript variant 1, mRNA.

2023


SNCA
NM_001146054

Homo sapiens synuclein, alpha (non A4 component of amyloid precursor) (SNCA), transcript variant 2, mRNA.

2024


SNCA
NM_001146055

Homo sapiens synuclein, alpha (non A4 component of amyloid precursor) (SNCA), transcript variant 3, mRNA.

2025


SNCA
NM_007308

Homo sapiens synuclein, alpha (non A4 component of amyloid precursor) (SNCA), transcript variant 4, mRNA.

2026


SNX10
NM_013322

Homo sapiens sorting nexin 10 (SNX10), transcript variant 2, mRNA.

2027


SNX10
NM_001199835

Homo sapiens sorting nexin 10 (SNX10), transcript variant 1, mRNA.

2028


SNX10
NM_001199837

Homo sapiens sorting nexin 10 (SNX10), transcript variant 3, mRNA.

2029


SNX10
NM_001199838

Homo sapiens sorting nexin 10 (SNX10), transcript variant 4, mRNA.

2030


SNX10
NR_037670

Homo sapiens sorting nexin 10 (SNX10), transcript variant 5, non-coding RNA.

2031


SP110
NM_004509

Homo sapiens SP110 nuclear body protein (SP110), transcript variant a, mRNA.

2032


SP110
NM_080424

Homo sapiens SP110 nuclear body protein (SP110), transcript variant c, mRNA.

2033


SP110
NM_001185015

Homo sapiens SP110 nuclear body protein (SP110), transcript variant d, mRNA.

2034


SP110
NM_004510

Homo sapiens SP110 nuclear body protein (SP110), transcript variant b, mRNA.

2035


SP140
NM_001005176

Homo sapiens SP140 nuclear body protein (SP140), transcript variant 2, mRNA.

2036


SP140
NM_007237

Homo sapiens SP140 nuclear body protein (SP140), transcript variant 1, mRNA.

2037


SPINK5
NM_001127698

Homo sapiens serine peptidase inhibitor, Kazal type 5 (SPINK5), transcript variant 1, mRNA.

2038


SPINK5
NM_006846

Homo sapiens serine peptidase inhibitor, Kazal type 5 (SPINK5), transcript variant 2, mRNA.

2039


SPINK5
NM_001127699

Homo sapiens serine peptidase inhibitor, Kazal type 5 (SPINK5), transcript variant 3, mRNA.

2040


SQSTM1
NM_003900

Homo sapiens sequestosome 1 (SQSTM1), transcript variant 1, mRNA.

2041


SQSTM1
NM_001142298

Homo sapiens sequestosome 1 (SQSTM1), transcript variant 2, mRNA.

2042


SQSTM1
NM_001142299

Homo sapiens sequestosome 1 (SQSTM1), transcript variant 3, mRNA.

2043


SRSF1
NM_001078166

Homo sapiens serine and arginine rich splicing factor 1 (SRSF1), transcript variant 2, mRNA.

2044


SRSF1
NM_006924

Homo sapiens serine and arginine rich splicing factor 1 (SRSF1), transcript variant 1, mRNA.

2045


SRSF1
NR_034041

Homo sapiens serine and arginine rich splicing factor 1 (SRSF1), transcript variant 3, non-coding RNA.

2046


STAT1
NM_007315

Homo sapiens signal transducer and activator of transcription 1 (STAT1), transcript variant alpha, mRNA.

2047


STAT1
NM_139266

Homo sapiens signal transducer and activator of transcription 1 (STAT1), transcript variant beta, mRNA.

2048


STAT2
NM_005419

Homo sapiens signal transducer and activator of transcription 2, 113 kDa (STAT2), transcript variant 1, mRNA.

2049


STAT2
NM_198332

Homo sapiens signal transducer and activator of transcription 2, 113 kDa (STAT2), transcript variant 2, mRNA.

2050


STAT3
NM_003150

Homo sapiens signal transducer and activator of transcription 3 (STAT3), transcript variant 2, mRNA.

2051


STAT3
NM_139276

Homo sapiens signal transducer and activator of transcription 3 (STAT3), transcript variant 1, mRNA.

2052


STAT3
NM_213662

Homo sapiens signal transducer and activator of transcription 3 (STAT3), transcript variant 3, mRNA.

2053


STAT5B
NM_012448

Homo sapiens signal transducer and activator of transcription 5B (STAT5B), mRNA.

2054


STIM1
NM_003156

Homo sapiens stromal interaction molecule 1 (STIM1), transcript variant 2, mRNA.

2055


STK4
NM_006282

Homo sapiens serine/threonine kinase 4 (STK4), mRNA.

2056


STX11
NM_003764

Homo sapiens syntaxin 11 (STX11), mRNA.

2057


STXBP2
NM_001127396

Homo sapiens syntaxin binding protein 2 (STXBP2), transcript variant 2, mRNA.

2058


STXBP2
NM_001272034

Homo sapiens syntaxin binding protein 2 (STXBP2), transcript variant 3, mRNA.

2059


STXBP2
NM_006949

Homo sapiens syntaxin binding protein 2 (STXBP2), transcript variant 1, mRNA.

2060


STXBP2
NR_073560

Homo sapiens syntaxin binding protein 2 (STXBP2), transcript variant 4, non-coding RNA.

2061


SYNCRIP
NM_001159673

Homo sapiens synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP), transcript variant 2, mRNA.

2062


SYNCRIP
NM_001159674

Homo sapiens synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP), transcript variant 3, mRNA.

2063


SYNCRIP
NM_001159676

Homo sapiens synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP), transcript variant 5, mRNA.

2064


SYNCRIP
NM_001159677

Homo sapiens synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP), transcript variant 6, mRNA.

2065


SYNCRIP
NM_001253771

Homo sapiens synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP), transcript variant 7, mRNA.

2066


SYNCRIP
NM_001159675

Homo sapiens synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP), transcript variant 4, mRNA.

2067


SYNCRIP
NM_006372

Homo sapiens synaptotagmin binding cytoplasmic RNA interacting protein (SYNCRIP), transcript variant 1, mRNA.

2068


T
NM_001270484

Homo sapiens T brachyury transcription factor (T), transcript variant 2, mRNA.

2069


T
NM_003181

Homo sapiens T brachyury transcription factor (T), transcript variant 1, mRNA.

2070


TAP1
NM_000593

Homo sapiens transporter 1, ATP binding cassette subfamily B member (TAP1), transcript variant 1, mRNA.

2071


TAP2
NM_018833

Homo sapiens transporter 2, ATP binding cassette subfamily B member (TAP2), transcript variant 2, mRNA.

2072


TAP2
NM_000544

Homo sapiens transporter 2, ATP binding cassette subfamily B member (TAP2), transcript variant 1, B allele, mRNA.

2073


TAPBP
NM_003190

Homo sapiens TAP binding protein (tapasin) (TAPBP), transcript variant 1, mRNA.

2074


TAPBP
NM_172209

Homo sapiens TAP binding protein (tapasin) (TAPBP), transcript variant 3, mRNA.

2075


TAPBP
NM_172208

Homo sapiens TAP binding protein (tapasin) (TAPBP), transcript variant 2, mRNA.

2076


TAZ
NM_000116

Homo sapiens tafazzin (TAZ), transcript variant 1, mRNA.

2077


TAZ
NM_181312

Homo sapiens tafazzin (TAZ), transcript variant 3, mRNA.

2078


TAZ
NM_181311

Homo sapiens tafazzin (TAZ), transcript variant 2, mRNA.

2079


TAZ
NM_181313

Homo sapiens tafazzin (TAZ), transcript variant 4, mRNA.

2080


TAZ
NR_024048

Homo sapiens tafazzin (TAZ), transcript variant 5, non-coding RNA.

2081


TBK1
NM_013254

Homo sapiens TANK binding kinase 1 (TBK1), mRNA.

2082


TBX1
NM_005992

Homo sapiens T-box 1 (TBX1), transcript variant B, mRNA.

2083


TBX1
NM_080646

Homo sapiens T-box 1 (TBX1), transcript variant A, mRNA.

2084


TBX1
NM_080647

Homo sapiens T-box 1 (TBX1), transcript variant C, mRNA.

2085


TCIRG1
NM_006019

Homo sapiens T-cell immune regulator 1, ATPase H+ transporting V0 subunit a3 (TCIRG1), transcript variant 1, mRNA.

2086


TCIRG1
NM_006053

Homo sapiens T-cell immune regulator 1, ATPase H+ transporting V0 subunit a3 (TCIRG1), transcript variant 2, mRNA.

2087


TICAM1
NM_182919

Homo sapiens toll like receptor adaptor molecule 1 (TICAM1), mRNA.

2088


TLR3
NM_003265

Homo sapiens toll like receptor 3 (TLR3), mRNA.

2089


TLR4
NM_003266

Homo sapiens toll like receptor 4 (TLR4), transcript variant 3, mRNA.

2090


TLR4
NM_138554

Homo sapiens toll like receptor 4 (TLR4), transcript variant 1, mRNA.

2091


TLR4
NM_138557

Homo sapiens toll like receptor 4 (TLR4), transcript variant 4, mRNA.

2092


TMEM173
NM_198282

Homo sapiens transmembrane protein 173 (TMEM173), mRNA.

2093


TNF
NM_000594

Homo sapiens tumor necrosis factor (TNF), mRNA.

2094


TNFAIP3
NM_001270507

Homo sapiens TNF alpha induced protein 3 (TNFAIP3), transcript variant 2, mRNA.

2095


TNFAIP3
NM_001270508

Homo sapiens TNF alpha induced protein 3 (TNFAIP3), transcript variant 1, mRNA.

2096


TNFAIP3
NM_006290

Homo sapiens TNF alpha induced protein 3 (TNFAIP3), transcript variant 3, mRNA.

2097


TNFRSF11A
NM_003839

Homo sapiens tumor necrosis factor receptor superfamily, member 11a, NFKB activator (TNFRSF11A), transcript variant 1, mRNA.

2098


TNFRSF11A
NM_001270949

Homo sapiens tumor necrosis factor receptor superfamily, member 11a, NFKB activator (TNFRSF11A), transcript variant 2, mRNA.

2099


TNFRSF11A
NM_001270950

Homo sapiens tumor necrosis factor receptor superfamily, member 11a, NFKB activator (TNFRSF11A), transcript variant 3, mRNA.

2100


TNFRSF11A
NM_001270951

Homo sapiens tumor necrosis factor receptor superfamily, member 11a, NFKB activator (TNFRSF11A), transcript variant 4, mRNA.

2101


TNFRSF11B
NM_002546

Homo sapiens tumor necrosis factor receptor superfamily, member 11b (TNFRSF11B), mRNA.

2102


TNFRSF13B
NM_012452

Homo sapiens TNF receptor superfamily member 13B (TNFRSF13B), mRNA.

2103


TNFRSF4
NM_003327

Homo sapiens TNF receptor superfamily member 4 (TNFRSF4), mRNA.

2104


TNFRSF8
NM_001243

Homo sapiens TNF receptor superfamily member 8 (TNFRSF8), transcript variant 1, mRNA.

2105


TNFSF11
NM_003701

Homo sapiens tumor necrosis factor (ligand) superfamily, member 11 (TNFSF11), transcript variant 1, mRNA.

2106


TNFSF11
NM_033012

Homo sapiens tumor necrosis factor (ligand) superfamily, member 11 (TNFSF11), transcript variant 2, mRNA.

2107


TNFSF12
NM_003809

Homo sapiens tumor necrosis factor superfamily member 12 (TNFSF12), transcript variant 1, mRNA.

2108


TNFSF12
NR_037146

Homo sapiens tumor necrosis factor superfamily member 12 (TNFSF12), transcript variant 2, non-coding RNA.

2109


TP53
NM_000546

Homo sapiens tumor protein p53 (TP53), transcript variant 1, mRNA.

2110


TP53
NM_001126112

Homo sapiens tumor protein p53 (TP53), transcript variant 2, mRNA.

2111


TP53
NM_001126113

Homo sapiens tumor protein p53 (TP53), transcript variant 4, mRNA.

2112


TP53
NM_001126114

Homo sapiens tumor protein p53 (TP53), transcript variant 3, mRNA.

2113


TP53
NM_001126115

Homo sapiens tumor protein p53 (TP53), transcript variant 5, mRNA.

2114


TP53
NM_001126116

Homo sapiens tumor protein p53 (TP53), transcript variant 6, mRNA.

2115


TP53
NM_001126117

Homo sapiens tumor protein p53 (TP53), transcript variant 7, mRNA.

2116


TP53
NM_001126118

Homo sapiens tumor protein p53 (TP53), transcript variant 8, mRNA.

2117


TP53
NM_001276695

Homo sapiens tumor protein p53 (TP53), transcript variant 4, mRNA.

2118


TP53
NM_001276696

Homo sapiens tumor protein p53 (TP53), transcript variant 3, mRNA.

2119


TP53
NM_001276697

Homo sapiens tumor protein p53 (TP53), transcript variant 5, mRNA.

2120


TP53
NM_001276698

Homo sapiens tumor protein p53 (TP53), transcript variant 6, mRNA.

2121


TP53
NM_001276699

Homo sapiens tumor protein p53 (TP53), transcript variant 7, mRNA.

2122


TP53
NM_001276760

Homo sapiens tumor protein p53 (TP53), transcript variant 1, mRNA.

2123


TP53
NM_001276761

Homo sapiens tumor protein p53 (TP53), transcript variant 2, mRNA.

2124


TRAF3
NM_001199427

Homo sapiens TNF receptor associated factor 3 (TRAF3), transcript variant 4, mRNA.

2125


TRAF3
NM_003300

Homo sapiens TNF receptor associated factor 3 (TRAF3), transcript variant 3, mRNA.

2126


TRAF3
NM_145725

Homo sapiens TNF receptor associated factor 3 (TRAF3), transcript variant 1, mRNA.

2127


TRAF3
NM_145726

Homo sapiens TNF receptor associated factor 3 (TRAF3), transcript variant 2, mRNA.

2128


TRAF6
NM_004620

Homo sapiens TNF receptor-associated factor 6, E3 ubiquitin protein ligase (TRAF6), transcript variant 2, mRNA.

2129


TRAF6
NM_145803

Homo sapiens TNF receptor-associated factor 6, E3 ubiquitin protein ligase (TRAF6), transcript variant 1, mRNA.

2130


TREX1
NM_007248

Homo sapiens three prime repair exonuclease 1 (TREX1), transcript variant 5, mRNA.

2131


TREX1
NM_033629

Homo sapiens three prime repair exonuclease 1 (TREX1), transcript variant 4, mRNA.

2132


TREX1
NM_016381

Homo sapiens three prime repair exonuclease 1 (TREX1), transcript variant 1, mRNA.

2133


TRNT1
NM_182916

Homo sapiens tRNA nucleotidyl transferase 1 (TRNT1), transcript variant 1, mRNA.

2134


TTC7A
NM_020458

Homo sapiens tetratricopeptide repeat domain 7A (TTC7A), transcript variant 2, mRNA.

2135


TYK2
NM_003331

Homo sapiens tyrosine kinase 2 (TYK2), mRNA.

2136


UNC119
NM_005148

Homo sapiens unc-119 lipid binding chaperone (UNC119), transcript variant 1, mRNA.

2137


UNC119
NM_054035

Homo sapiens unc-119 lipid binding chaperone (UNC119), transcript variant 2, mRNA.

2138


UNC13D
NM_199242

Homo sapiens unc-13 homolog D (UNC13D), mRNA.

2139


UNC93B1
NM_030930

Homo sapiens unc-93 homolog B1 (C. elegans) (UNC93B1), mRNA.

2140


UNG
NM_080911

Homo sapiens uracil DNA glycosylase (UNG), transcript variant 2, mRNA.

2141


UNG
NM_003362

Homo sapiens uracil DNA glycosylase (UNG), transcript variant 1, mRNA.

2142


USP18
NM_017414

Homo sapiens ubiquitin specific peptidase 18 (USP18), mRNA.

2143


USP20
NM_006676

Homo sapiens ubiquitin specific peptidase 20 (USP20), transcript variant 1, mRNA.

2144


USP20
NM_001008563

Homo sapiens ubiquitin specific peptidase 20 (USP20), transcript variant 2, mRNA.

2145


USP20
NM_001110303

Homo sapiens ubiquitin specific peptidase 20 (USP20), transcript variant 3, mRNA.

2146


VAPA
NM_003574

Homo sapiens VAMP associated protein A (VAPA), transcript variant 1, mRNA.

2147


VAPA
NM_194434

Homo sapiens VAMP associated protein A (VAPA), transcript variant 2, mRNA.

2148


VCP
NM_007126

Homo sapiens valosin containing protein (VCP), mRNA.

2149


VDAC1
NM_003374

Homo sapiens voltage dependent anion channel 1 (VDAC1), transcript variant 1, mRNA.

2150


VDAC1
NR_036624

Homo sapiens voltage dependent anion channel 1 (VDAC1), transcript variant 3, non-coding RNA.

2151


VDAC1
NR_036625

Homo sapiens voltage dependent anion channel 1 (VDAC1), transcript variant 2, non-coding RNA.

2152


VPS13B
NM_017890

Homo sapiens vacuolar protein sorting 13 homolog B (yeast) (VPS13B), transcript variant 5, mRNA.

2153


VPS13B
NM_181661

Homo sapiens vacuolar protein sorting 13 homolog B (yeast) (VPS13B), transcript variant 4, mRNA.

2154


VPS13B
NM_015243

Homo sapiens vacuolar protein sorting 13 homolog B (yeast) (VPS13B), transcript variant 3, mRNA.

2155


VPS13B
NR_047582

Homo sapiens vacuolar protein sorting 13 homolog B (yeast) (VPS13B), transcript variant 6, non-coding RNA.

2156


VPS13B
NM_152564

Homo sapiens vacuolar protein sorting 13 homolog B (yeast) (VPS13B), transcript variant 1, mRNA.

2157


VPS45
NM_007259

Homo sapiens vacuolar protein sorting 45 homolog (VPS45), transcript variant 1, mRNA.

2158


WAS
NM_000377

Homo sapiens Wiskott-Aldrich syndrome (WAS), mRNA.

2159


WEE1
NM_003390

Homo sapiens WEE1 G2 checkpoint kinase (WEE1), transcript variant 1, mRNA.

2160


WEE1
NM_001143976

Homo sapiens WEE1 G2 checkpoint kinase (WEE1), transcript variant 2, mRNA.

2161


WIPF1
NM_001077269

Homo sapiens WAS/WASL interacting protein family member 1 (WIPF1), transcript variant 2, mRNA.

2162


WIPF1
NM_003387

Homo sapiens WAS/WASL interacting protein family member 1 (WIPF1), transcript variant 1, mRNA.

2163


XIAP
NM_001204401

Homo sapiens X-linked inhibitor of apoptosis, E3 ubiquitin protein ligase (XIAP), transcript variant 2, mRNA.

2164


XIAP
NM_001167

Homo sapiens X-linked inhibitor of apoptosis, E3 ubiquitin protein ligase (XIAP), transcript variant 1, mRNA.

2165


XIAP
NR_037916

Homo sapiens X-linked inhibitor of apoptosis, E3 ubiquitin protein ligase (XIAP), transcript variant 3, non-coding RNA.

2166


YBX1
NM_004559

Homo sapiens Y-box binding protein 1 (YBX1), transcript variant 1, mRNA.

2167


YWHAZ
NM_001135699

Homo sapiens tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ), transcript variant 3, mRNA.

2168


YWHAZ
NM_001135700

Homo sapiens tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ), transcript variant 4, mRNA.

2169


YWHAZ
NM_001135701

Homo sapiens tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ), transcript variant 5, mRNA.

2170


YWHAZ
NM_001135702

Homo sapiens tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ), transcript variant 6, mRNA.

2171


YWHAZ
NM_003406

Homo sapiens tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ), transcript variant 1, mRNA.

2172


YWHAZ
NM_145690

Homo sapiens tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ), transcript variant 2, mRNA.

2173


ZAP70
NM_001079

Homo sapiens zeta chain of T cell receptor associated protein kinase 70 (ZAP70), transcript variant 1, mRNA.

2174


ZAP70
NM_207519

Homo sapiens zeta chain of T cell receptor associated protein kinase 70 (ZAP70), transcript variant 2, mRNA.

2175


ZBTB24
NM_014797

Homo sapiens zinc finger and BTB domain containing 24 (ZBTB24), transcript variant 1, mRNA.

2176


ZBTB24
NM_001164313

Homo sapiens zinc finger and BTB domain containing 24 (ZBTB24), transcript variant 2, mRNA.

2177









Table 12 lists all transcript variants for genes in Table 6 that were not ‘discovered’ by PBio on the basis of aCGH (CNV identified genes). The SEQ ID NOs correspond to transcript variants (oftentimes more than one per gene).









TABLE 13







Genes for which the total burden of heterozygous, damaging variants was


found to be statistically greater in PML cases versus ExAC controls
















Ave
Ave EXAC
Ave EXAC

Ave FET





GENE
CASES
CASES
SAMPLES
Ave FET
corr(419)
Ave OR
Ethnicity
Overlap


















PLCG2
17
1,806
31,277
1.43E−10
6.21E−08
10.27
EUR
EUR + AFR


RBCK1
6
187
29,324
4.27E−07
1.86E−04
24.60
EUR



EPG5
9
764
32,835
7.11E−07
3.09E−04
10.79
EUR



IL17F
4
61
33,346
1.67E−06
7.28E−04
54.57
EUR



SHARPIN
8
646
32,162
2.58E−06
1.12E−03
10.84
EUR



PRF1
8
715
33,027
4.44E−06
1.93E−03
10.04
EUR



JAGN1
5
163
27,768
6.80E−06
2.96E−03
21.71
EUR



TAP1
5
203
28,125
1.80E−05
7.82E−03
17.63
EUR



POLE
11
1,660
29,108
2.84E−05
1.23E−02
5.51
EUR
EUR + AFR


LRBA
11
1,876
32,136
3.47E−05
1.51E−02
5.38
EUR



EHF
3
49
32,588
4.83E−05
2.10E−02
48.59
EUR



IL12B
3
58
33,112
7.44E−05
3.23E−02
41.70
EUR



ATL2
8
31
5,041
4.03E−11
1.75E−08
90.11
AFR



NHEJ1
6
27
4,384
5.48E−09
2.39E−06
64.56
AFR



LYST
11
291
4,748
1.09E−08
4.76E−06
16.85
AFR



HIVEP1
9
150
4,432
7.41E−08
3.22E−05
23.83
AFR



AP3B1
5
46
4,937
1.69E−06
7.36E−04
33.23
AFR



TNFRSF10A
7
149
4,626
3.28E−06
1.43E−03
15.03
AFR



PIK3CD
7
148
4,549
3.52E−06
1.53E−03
14.87
AFR



PLCG2
8
256
4,410
1.47E−05
6.41E−03
9.99
AFR
EUR + AFR


PNP
3
11
5,189
2.00E−05
8.69E−03
78.45
AFR



POLE
8
297
4,752
2.48E−05
1.08E−02
9.23
AFR
EUR + AFR


MCEE
3
13
5,164
3.10E−05
1.35E−02
66.04
AFR



DOCK2
6
173
5,023
6.39E−05
2.78E−02
11.21
AFR



ALG12
4
43
4,252
6.73E−05
2.93E−02
23.03
AFR









Table 13 lists genes for which the total burden of heterozygous, damaging variants was found to be statistically greater in PML cases versus ExAC controls. Gene burden analysis was performed as described below at minor allele frequency (MAF) cutoffs of 0.01, 0.02, 0.03, 0.04 and 0.05. Not all genes survived statistical analysis at all MAF cutoffs. For each gene that survived at multiple MAF cutoffs, the averages of the Fishers Exact Test (FET), nominal and corrected, were calculated, as were the other relevant metrics. Two genes overlapped between AFR and EUR analyses. FETs were corrected for multiple testing with the number of genes used in this study (419). Only genes for which FET_Corr was <0.05 and in which variants affected >10% of cases within the given ethnicity (>2 for AFR, >4 for EUR) were considered for inclusion.









TABLE 14





Top tier of variants found to be significant on the basis of variant burden analysis

























PML
PML
PML







EUR
AFR
LAT
ExAC
ExAC


Gene
Variant (hg19)
Genotype
44
21
5
EUR
AFR





PLCG2
chr16: 81942175,
het
2
5
0
 512/32281
88/4707



A > G








IFIH1
chr2: 163136505,
het
6
1
0
 611/33155
23/5182



C > G








TCIRG1
chr11: 67818269,
het
0
4
0
 103/33193
200/5170



G > A








IGLL1
chr22: 23917192,
het
4
3
1
 751/33348
603/5183



G > T








MAVS
chr20: 3846397,
hom
4
4
0
 800/32122
684/4982



C > T








SHARPIN
chr8: 145154222,
het
8
4
0
2916/33177
59/4865



G > A








CHD7
chr8: 61654298,
het
5
0
0
1103/33106
39/4840



T > A








CX3CR1
chr3: 39323163,
hom
11
4
0
4723/31219
193/4376



A > C








LRBA
chr4: 151199080,
hom
3
3
0
2260/33328
20/5195



G > A








HIVEP3
chr1: 42047208,
het
5
3
1
3383/32494
123/5061



C > G








IFIH1
chr2: 163124051,
hom
20
3
1
12107/33356 
184/5199



C > T








RNASEL
chr1: 182554557,
hom
7
2
0
4543/33356
78/5202



C > T



















PML
PML
PML
PML
PML
PML



ExAC
EUR
EUR
AFR
AFR
ALL
ALL


Gene
LAT
OR
FET
OR
FET
OR
FET





PLCG2
116/5548
2.95
0.154755
16.40
0.0000
6.49
0.0002


IFIH1
119/5671
8.41
0.000156
11.22
0.0927
6.38
0.0002


TCIRG1
 60/5770
NA
NA
5.85
0.0082
7.31
0.0028


IGLL1
236/5782
4.34
0.017218
1.27
0.7286
3.47
0.0036


MAVS
 52/5691
3.92
0.023868
1.48
0.5191
3.47
0.0036


SHARPIN
171/5780
2.31
0.053526
19.17
0.0001
2.68
0.0040


CHD7
 93/5725
3.72
0.015268
NA
NA
2.64
0.0485


CX3CR1
1357/5491 
1.87
0.088087
5.10
0.0128
1.51
0.1806


LRBA
 54/5785
1.01
1.000000
43.13
0.0001
1.69
0.2736


HIVEP3
902/5756
1.10
0.803620
6.69
0.0143
1.30
0.4283


IFIH1
1076/5776 
1.46
0.212471
4.54
0.0374
1.21
0.4372


RNASEL
167/5785
1.20
0.658473
6.91
0.0403
1.22
0.5616









Table 14 lists the top tier of variants that were found to be significant on the basis of variant burden analysis, as described below. For each variant (genome coordinates are based on UCSC hg19), detailed information is presented of the numbers of EUR and AFR cases that carry the variant, along with the ethnic-specific and aggregate statistical metrics.









TABLE 15





Second tier of variants found on the basis of variant burden analysis

























PML
PML
PML







EUR
AFR
LAT
ExAC
ExAC


Gene
Variant (hg19)
Genotype
44
21
5
EUR
AFR





SHARPIN
chr8: 145154824,
het
3
0
0
 2/30,670
 0/4,471



A > C








RTEL1
chr20: 62305450,
het
0
2
0
 1/32,552
 0/4,838



C > T








IGLL1
chr22: 23915745,
het
2
0
1
 19/33,348
74/5,184



G > A








PGM3
chr6: 83884161,
het
0
2
0
 0/33,069
26/5,167



C > G








ATM
chr11: 108202772,
het
3
0
0
170/32,707
 3/5,099



G > T








TMEM173
chr5: 138856923,
het
2
2
0
108/32,327
204/4,842 



C > T








CLCN7
chr16: 1510535,
het
0
2
0
 1/32,898
66/5,119



C > T








MAVS
chr20: 3843027,
hom
4
2
0
803/33,206
167/5,171 



C > A








ORAI1
chr12: 122064788,
het
4
0
0
371/28,708
 5/3,555



G > GT








RBFOX1
chr16: 7714909,
het
0
2
0
 1/33,367
69/4,902



C > T








MALT1
chr18: 56401523,
het
4
0
0
466/33,239
 9/5,179



C > T








GFI1
chr1: 92946625,
het
2
1
0
206/29,111
 6/4,156



G > C








DOCK2
chr5: 169081453,
het
0
2
0
 48/33.350
43/5,201



G > C








ATM
chr11: 108117787,
het
2
0
0
 93/33,256
 2/5,151



C > T








SNAP29
chr22: 21235389,
het
3
0
0
283/32,917
21/5,149



A > G








TICAM1
chr19: 4817657,
het
0
2
0
 32/31,437
71/4,814



C > T








GTPBP4
chr10: 1060218,
hom
3
0
0
334/33,367
20/5,202



G > A








BACH1
chr21: 30698953,
het
2
0
0
134/33,122
 4/5,103



T > G








DOCK8
chr9: 304628,
het
2
0
0
149/33,298
 2/5,161



G > A








STXBP2
chr19: 7712287,
het
2
0
0
161/32,104
 4/4,626



G > C








FAS
chr10: 90771767,
het
2
0
0
175/33,304
 3/5,182



G > A








GOLGB1
chr3: 121415370,
het
3
2
0
1,111/33,349
26/5,179



T > C








FUK
chr16: 70503095,
het
4
0
0
741/33,341
23/4,899



A > G








IL10
chr1: 206945738,
het
2
0
0
206/33,343
 2/5,198



C > T








ITK
chr5: 156593120,
het
2
0
0
206/33,353
 5/5,203



C > T








STIM2
chr4: 27019452,
het
2
0
0
219/33,369
 5/5,202



C > T








ASH1L
chr1: 155317682,
het
2
0
0
218/33,367
 2/5,203



C > T








TBC1D16
chr17: 77926526,
het
3
0
0
496/31,905
15/4,845



C > T








LYST
chr1: 235840495,
het
3
0
0
517/33,239
10/5,156



G > T








SALL2
chr14: 21993359,
het
3
0
0
519/31,729
14/4,520



G > A








CHD7
chr8: 61757805,
het
3
0
0
517/32,880
14/4,872



C > T








BLM
chr15: 91306241,
het
2
0
0
266/33,277
10/5,061



G > A








NOD2
chr16: 50741791,
het
0
2
0
285/33,369
21/5,203



C > T








IGLL1
chr22: 23915583,
het
2
0
0
265/33,334
21/5,183



T > C








TTC7A
chr2: 47205921,
het
3
0
0
589/33,202
13/5,173



C > T








KITLG
chr12: 88900891,
het
4
0
0
1,023/33,226
31/5,158



C > A








ATR
chr3: 142281353,
het
4
0
0
1,037/33,343
14/5,130



C > G








ATM
chr11: 108123551,
het
0
2
0
217/29,921
66/4,955



C > T








CR2
chr1: 207641950,
het
0
2
0
391/33,363
19/5,203



C > T








HIVEP2
chr6: 143092151,
het
3
2
0
1,718/33,370
50/4,901



T > C








ITSN2
chr2: 24431184,
hom
3
2
0
2,019/33,339
17/5,186



C > T








ITSN2
chr2: 24432937,
hom
3
2
0
2,026/32,472
17/4,958



C > T








DOCK8
chr9: 312134,
het
3
2
0
2,114/33,251
79/5,180



G > A








VPS13B
chr8: 100205255,
het
0
2
0
811/33,345
19/5,192



G > A








NRIP1
chr21: 16339852,
het
0
2
0
901/33,355
19/5,203



T > C



















PML
PML
PML
PML
PML
PML



ExAC
EUR
EUR
AFR
AFR
ALL
ALL


Gene
LAT
OR
FET
OR
FET
OR
FET





SHARPIN
 0/5,302
1122.00  
0.000000
NA
NA
905.40
0.0000


RTEL1
 0/5,737
NA
NA
1240.64 
0.0000
1268.41
0.0000


IGLL1
 9/5,783
83.53 
0.000351
NA
NA
19.41
0.0006


PGM3
 3/5,748
NA
NA
20.81
0.0055
44.58
0.0011


ATM
 7/5,713
14.00 
0.001636
NA
NA
10.78
0.0032


TMEM173
58/5,770
14.21 
0.009863
 2.39
0.2226
6.97
0.0033


CLCN7
 0/5,732
NA
NA
 8.06
0.0308
19.18
0.0055


MAVS
46/5,779
4.04
0.021706
 3.15
0.1480
3.98
0.0056


ORAI1
16/5,354
7.64
0.002562
NA
NA
5.76
0.0064


RBFOX1
 4/5,782
NA
NA
 7.37
0.0361
17.48
0.0066


MALT1
40/5,760
7.03
0.003411
NA
NA
5.14
0.0093


GFI1
39/5,114
6.68
0.039391
34.58
0.0347
6.80
0.0113


DOCK2
27/5,786
NA
NA
12.63
0.0137
11.02
0.0155


ATM
28/5,756
16.98 
0.007047
NA
NA
10.53
0.0169


SNAP29
32/5,740
8.44
0.006584
NA
NA
5.79
0.0171


TICAM1
19/5,687
NA
NA
 7.03
0.0392
10.08
0.0183


GTPBP4
21/5,786
7.24
0.009925
NA
NA
5.25
0.0220


BACH1
 6/5,778
11.72 
0.014110
NA
NA
8.96
0.0227


DOCK8
 5/5,762
10.59 
0.017020
NA
NA
8.31
0.0261


STXBP2
11/5,686
9.45
0.021028
NA
NA
7.06
0.0350


FAS
10/5,731
9.01
0.022902
NA
NA
6.89
0.0365


GOLGB1
84/5,779
2.12
0.180743
20.86
0.0055
2.71
0.0443


FUK
73/5,787
4.40
0.016488
NA
NA
3.13
0.0449


IL10
 6/5,787
7.66
0.030787
NA
NA
6.06
0.0458


ITK
 5/5,789
7.66
0.030770
NA
NA
6.01
0.0466


STIM2
 9/5,789
7.21
0.034346
NA
NA
5.57
0.0532


ASH1L
17/5,789
7.24
0.034067
NA
NA
5.48
0.0548


TBC1D16
21/5,707
4.63
0.031269
NA
NA
3.53
0.0584


LYST
35/5,756
4.63
0.031299
NA
NA
3.47
0.0606


SALL2
17/5,718
4.40
0.035528
NA
NA
3.37
0.0650


CHD7
41/5,765
4.58
0.032169
NA
NA
3.36
0.0654


BLM
17/5,756
5.91
0.048875
NA
NA
4.40
0.0799


NOD2
 2/5,789
NA
NA
25.97
0.0037
4.21
0.0860


IGLL1
26/5,787
5.94
0.048403
NA
NA
4.15
0.0881


TTC7A
61/5,759
4.05
0.043427
NA
NA
2.94
0.0891


KITLG
40/5,760
3.15
0.046242
NA
NA
2.38
0.0964


ATR
69/5,785
3.12
0.047671
NA
NA
2.33
0.1021


ATM
40/5,425
NA
NA
 7.80
0.0327
3.64
0.1093


CR2
 8/5,754
NA
NA
28.72
0.0031
3.09
0.1422


HIVEP2
209/5,788 
1.35
0.494339
10.21
0.0202
1.64
0.2458


ITSN2
55/5,784
1.14
0.748301
32.01
0.0025
1.55
0.3862


ITSN2
56/5,672
1.10
0.753875
30.59
0.0028
1.50
0.3937


DOCK8
161/5,768 
1.08
0.757661
 6.80
0.0415
1.37
0.4238


VPS13B
100/5,778 
NA
NA
28.66
0.0031
1.37
0.6600


NRIP1
64/5,780
NA
NA
28.72
0.0031
1.30
0.6698









Table 15 lists the second tier of variants that were found on the basis of variant burden analysis, as described below. For each variant (genome coordinates are UCSC hg19), detailed information is presented of the numbers of EUR and AFR cases that carry the variant, along with the ethnic-specific and aggregate statistical metrics.









TABLE 16







Potential testing scenario, based on top variant burden hits











Proportion

Patient information














Cases
of Cohort
Test
Primary




Gene/Variant
solved
(n = 70)
Method
disease
Ethnicity
Gender
















All 4 SNVs
28
40%
genotyping
M, H, O
A, E
both


SHARPIN, IFIH1,
24
34%
genotyping
M, H, O
A, E
both


PLCG2 SNVs








IFIH1, PLCG2 SNVs
13
19%
genotyping
M, H, O
A, E
both


SHARPIN SNV
13
19%
genotyping
M, H
A, E
both


IFIH1 SNV
7
10%
genotyping
M, H, O
A, E
both


PLCG2 SNV
7
10%
genotyping
M, H
A, E
both


CHD7 SNV
5
 7%
genotyping
M, H, O
E
both









Table 16 lists a potential testing scenario, based on top variant burden hits (reported in Table 14). The analysis is for illustrative purposes only, it being acknowledged that greater diagnostic yields can be obtained by assaying for a larger number of variants, including those listed in Table 15. Examples are given for diagnostic yield using singleton variants, as well as a variety of combinations, including the use of the top 4 variants. For this set of variants, the test method is described as genotyping, as opposed to whole gene sequencing (i.e., determination of the status at each of the bases, which yields a binary output, as opposed to identification of variants elsewhere in the relevant genes).









TABLE 17







Potential testing scenario using genes identified as having a greater


burden of damaging, heterozygous variants in the PML cohort
















Overall






Ethnic-
yield




Ave
Eth-
specific
(EUR +
Test


GENE
CASES
nicity
yield (%)
AFR) (%)
Method















PLCG2
17/44
EUR
38
38
Gene sequencing


PLCG2
 8/21
AFR
38

Gene sequencing


POLE
 8/21
AFR
38

Gene sequencing


POLE
11/44
EUR
25
29
Gene sequencing


LRBA
11/44
EUR
25

Gene sequencing


EPG5
 9/44
EUR
20

Gene sequencing


SHARPIN
 8/44
EUR
18

Gene sequencing









Table 17 lists a potential testing scenario using genes identified as having a greater burden of damaging, heterozygous variants in the PML cohort (see Table 13). The nature of the testing method is ‘gene sequencing’ since the variants are not known in advance—any and all potentially damaging variants need to be considered in such an assay.









TABLE 18







Summary of genes that survive case-level,


gene burden and/or variant burden analyses












Gene
Case Level
Variant Burden
Gene Burden






PLCG2
Yes
Yes
Yes



CHD7
Yes
Yes




IFIH1
Yes
Yes




AP3B1
Yes

Yes



EPG5
Yes

Yes



PIK3CD
Yes

Yes



LRBA

Yes
Yes



SHARPIN

Yes
Yes









Table 18 represents a summary of genes that survive case-level (2 or more examples in Tables 7, 8), gene burden and/or variant burden analyses (based on Tables 13 and 14). Of note is that PLCG2 satisfies all 3 criteria (2 or more examples, in Table 8, presence in Tables 13, 14). This summary demonstrates that many genes have been identified as significant on the basis of independent analysis methods.


Example 11—Figures Referenced in this Study


FIGS. 1-12 represent example CNV data from the PML gene discovery study (71 PML cases, see Table 7 for patient information) using array CGH (methods described herein). In each figure/drawing: 1) genome coordinates are listed at the top (hg18 assembly, chromosome number and position depicted); 2) data track 1 (labeled ‘Genes’) depicts the location of the RefSeq genes (exons are dark gray portions of the bars, introns are light gray portions of the bars); 3) data track 2 (labeled ‘Normal Cohort’) depicts the size and location of CNVs found in the NVE cohort (PBio's proprietary control database consisting of CNV findings in apparently healthy—i.e. normal—subjects, see methods herein) with the y-axis corresponding to the number of NVE subjects that have the CNV; and 4) remaining data tracks are CNV data found in individual PML patients wherein the y-axis corresponds to the log 2 ratio (see methods herein), points represent individual probes on the microarray, and line segments are shifted positive (copy number gain) or negative (copy number loss) based on the output of DNAcopy, the CNV calling algorithm. Typical log 2 ratios for gains and losses on the Agilent 1M microarray (see methods herein) and our experimental protocols are: 0.6 for duplications, 1.0 for triplications (or homozygous duplications), −1.0 for heterozygous deletions, and <−2 (often −4.0 to −6.0) for homozygous deletions. Relevant genes are labeled in the ‘Genes’ data track.



FIG. 1 represents an example of a gene impacted by germline and acquired CNVs. Germline CNVs that impact the PRKCB gene include patient PML50 with a 4.8 Kb intronic heterozygous loss (also found in 7 Normal subjects) and patient PML11 with a 7.3 Kb intronic gain (also found in 1 Normal subject). Acquired CNVs were found in 6 PML patients, a series of gains at −23.9 Mb with varying log 2 ratios, suggestive of a mixed cell population (array CGH experiments were performed on blood-derived genomic DNA).



FIG. 2 represents an example of potentially PML-relevant genes (TNFRSF13C and CENPM) impacted by acquired CNVs. Acquired CNVs were found in 9 PML patients, a series of gains at −40.6 Mb with varying log 2 ratios, suggestive of a mixed cell population (array CGH experiments were performed on blood-derived genomic DNA). All 9 PML patients (see Table 7 for patient information) had a primary diagnosis of HIV and were mixed gender (3 females and 6 males) and ethnicity (4 African ancestry and 5 European ancestry).



FIG. 3 represents an example of a gene impacted by germline and acquired CNVs. A germline CNV, which is a 7.2 Kb intronic heterozygous loss (not found in Normal subjects, but an adjacent loss is found in 8 Normal subjects) that impacts the PKHD1 gene, was detected in patient PML26. Acquired CNVs were found in 3 PML patients, a series of gains at −51.9 Mb with varying log 2 ratios, suggestive of a mixed cell population (array CGH experiments were performed on blood-derived genomic DNA).



FIG. 4 represents an example of a gene impacted by a recurrent CNV loss. The 14.7 Kb intronic deletion impacts the BMPR2 gene. Heterozygous deletions were detected in patients PML58 and MVGS811-13a (also found in 2 Normal subjects), and a homozygous deletion was detected in patient PML29 (none found in Normal subjects). All three PML patients are males and their primary disease is HIV (see Table 7).



FIG. 5 represents an example of a gene impacted by a recurrent CNV gain. The 10.2 Kb exonic gain disrupts the COMMD6 gene. Two PML patients, PML29 and MVGS811-13a, have a homozygous duplication (log 2 ratio comparable to triplications) based on the observation that 1000 genomes subjects are reported to have this gain (see hg19 assembly DGV variant esv3632749, which reports 148 of 2504 subjects as having this gain; no Normals were found in PBio's NVE db). Both PML patients are males and their primary disease is HW (see Table 7).



FIG. 6 represents an example of a gene impacted by a recurrent CNV gain. The 27.4 Kb exonic gain disrupts the KCTD7 gene and the right breakpoint is 16-90 Kb upstream of RABGEF1 transcript variants (RefSeq: NM_001287060, NR 104676, NM_014504, NM_001287062, NM_001287061). Patient PML29 has a homozygous duplication (log 2 ratio comparable to triplications) based on the observation that 1000 genomes subjects are reported to have this gain (see hg19 assembly DGV variant esv3613515, which reports 28 of 2504 subjects as having this gain; no Normals were found in PBio's NVE db). Patient PML63 has a duplication. Both PML patients are males of African ancestry and their primary disease is HIV (see Table 7).



FIG. 7 represents an example of a gene impacted by a recurrent CNV gain. The 344 Kb exonic gain disrupts the FPR2 and ZNF616 genes (via left and right breakpoints) and additional genes fully encompassed by this CNV are: FPR3, ZNF350, ZNF350-AS1, ZNF432, ZNF577, ZNF613, ZNF614, ZNF615, ZNF649, ZNF649-AS1, ZNF841. Patient PML03 has a homozygous duplication (log 2 ratio comparable to triplications) based on the observation that 3 Normal subjects (PBio's NVE db) are found to have a duplication of this region, along with patient PML10. Both PML patients are females of European ancestry and their primary diseases are HIV and MS (see Table 7).



FIG. 8 represents an example of a gene impacted by a recurrent CNV loss. The 1.1 Kb exonic deletion impacts the PIK3CD and PIK3CD-AS1 (previous gene symbol was C1orf200) genes. A homozygous deletion was detected in patient MVGS811-13a and this loss (heterozygous or homozygous) was not found in Normal subjects or the DGV public CNV database. The PML patient is a male and his primary disease is HIV (see Table 7). He is presumed to be of EUR ancestry (ethnicities were not available for MVGS samples).



FIG. 9 represents an example of a gene impacted by an intergenic, recurrent CNV gain. The 16.7 Kb intergenic gain has a left breakpoint that is 105 Kb upstream of the CD180 gene (RefSeq transcript variant NM 005582). Patient MVGS995-4a has a homozygous duplication (log 2 ratio comparable to triplications) based on the observation that 1000 genomes subjects are reported to have this gain (see hg19 assembly DGV variant esv3605336, which reports 2 of 2504 subjects as having this gain; no Normals were found in PBio's NVE db). The PML patient is a male of European ancestry and his primary disease is MS (see Table 7).



FIG. 10 represents an example of a gene impacted by an intergenic, recurrent CNV loss. The 7.7 Kb intergenic homozygous deletion has a left breakpoint that is 3-4 Kb upstream of VDAC1 transcript variants (RefSeq: NM_003374, NR_036625, NR_036624). This loss (heterozygous or homozygous) was not found in Normal subjects or the DGV public CNV database. Patient PML30 is a male of European ancestry and his primary disease is HW (see Table 7).



FIG. 11 represents an example of a gene impacted by an intergenic, recurrent CNV loss. The 6.8 Kb intergenic homozygous deletion has a left breakpoint that is 4 Kb downstream of EGR1 transcript variant (RefSeq: NM_001964) and 26 Kb downstream of ETF1 transcript variants (RefSeq: NM 001256302, NM_004730, NM_001282185, NM_001291975, NM_001291974). This loss was found to be homozygous in 1 Normal subject and the loss was also reported in the DGV public CNV database (see hg19 assembly DGV variant esv3606925, which reports 33 of 2504 subjects as having this loss, homozygous vs. heterozygous subjects are unknown). Patient PML69 is a male of European ancestry and his primary disease (condition) is kidney transplant (see Table 7, reported as ‘Other’). Patient PML69 was treated with CTLA4-Ig (belatacept, a CD28-B7 costimulation blocker and T-cell anergy inducer). The CD28 pathway includes links to the patient's genetic finding (e.g., homozygous deletion adjacent to the EGR1 gene) and several other genes that may be related to immunodeficiency (e.g., CD40LG, ITK, LCK, LRBA, PIK3CD, PIK3R1, PLCG2, WAS, and ZAP70) (Dekeyser M et al. Open Forum Infect Diseases, 2016, Refractory T-Cell Anergy and Rapidly Fatal Progressive Multifocal Leukoencephalopathy following Prolonged CTLA4 Therapy).



FIG. 12 represents an example of a gene impacted by an intergenic, recurrent CNV loss. The 5.6 Kb intergenic homozygous deletion has a left breakpoint that is 20 Kb upstream of ITSN2 transcript variants (RefSeq: NM_019595, NM_006277, NM_147152). Heterozygous losses were found in 50 Normal subjects and the loss was also reported in the DGV public CNV database (see hg19 assembly DGV variant esv3590068, which reports 222 of 2504 subjects as having this loss, homozygous vs. heterozygous subjects are unknown). Patient PML65 is a male of African ancestry and his primary disease is HIV (see Table 7).



FIG. 13 represents an example of known and/or predicted protein interactions using the String database (string-db.org; see Szklarczyk et al., (2015) and references therein). A non-redundant list of all genes reported in Table 7 (43 genes, which included those whose expression was inferred to be impacted by a nearby intergenic CNV) as best solutions/explanations for 61 of 71 PML cases (11 PML cases are reported as ‘unsolved’, including 1 case for which only CGH data was obtained) was assessed using the String db. The ‘minimum required interaction score’ was set to ‘high confidence (0.7)’ and no additional ‘interactors’ were added. Of the 43 input genes, 21 were found to have high confidence interactions, as shown in the figure, along with annotation of the number of PML cases that had each of these genes as a solution/explanation (e.g., 3 PML cases in Table 7 were found to have a PLCG2 solution).


Example 12—Gene Burden Analysis

Gene burden analysis was performed as follows. Using a variety of in-house scripts, and data downloaded from ExAC (exac.broadinstitute.org), a count was performed for all variants occurring in each of the 419 genes listed in Table 6. Each variant was classified according to whether it was deemed damaging (on the basis of at least one of the prediction algorithms SIFT, PolyPhen2 or MutationTaster) or non-damaging, heterozygous or homozygous. This was performed in parallel for PML variants and those found in ExAC. ExAC data for which quality/coverage was <80% of expected was not used and gene burden analysis could not therefore be performed.


An ethnic-specific (EUR or AFR only, there were too few LAT cases for this type of analysis) comparison was then performed for each of 4 categories:

    • Homozygous damaging
    • Homozygous non-damaging
    • Heterozygous damaging
    • Heterozygous non-damaging


For all 4 categories, variants with minor allele frequency (MAF) cutoffs of 0.01, 0.02. 0.03. 0.04, 0.05 and 0.1 were considered.


For each comparison, odds ratios (OR) and Fisher's exact test (FET) were calculated for the comparison of numbers of PML cases with at least one variant of the type under consideration and those in ExAC. Correction for multiple testing was performed by multiplying the FET by the number of genes being considered (419). Only genes for whom the FET_corrected was <0.05 were included in Table 13, which contains data on the average values for a given gene at all MAFs that passed FET_correction. In practice, only the category of heterozygous damaging yielded significant genes.


Example 13—Variant Burden Analysis

For each variant identified in at least one PML case, a count was performed in order to obtain the frequency of the same variant in the cohort as a whole. This aggregate data was compared to counts for the same variant as reported in ExAC. ExAC data was filtered for quality/coverage and variant burden analysis was not performed if ExAC coverage was <80% expected.


Variant burden analysis was performed separately for EUR (n=44 cases) and AFR (n=21 cases) cohorts (LAT cohort was too small) and the OR and FET values calculated. From this analysis, only variants with OR>1 (i.e., potentially indicative of increased risk for PML) for both ethnicities (AFR and EUR) and for which the ExAC frequency of the variant was <5% were considered. Furthermore, only those variants for which the frequency in the ethnic-specific cohort was >10% (5 or more EUR cases, 3 or more AFR cases) were considered top-tier (Table 14), although other variants have been tabulated in Table 15.


Example 14— Exemplary PML Risk Prediction Tests

Table 16 provides exemplary markers for creating a low-cost, simple (genotype specific SNVs) PML risk prediction test. Other embodiments could be similarly devised from other SNVs reported in Tables 14 and 15. Different combinations of SNVs from Tables 14, 15 could be utilized in tests of varying complexity, to develop a test that would yield higher diagnostic yields than the top example listed in Table 16 (i.e., 40%).


Table 17 provides exemplary genes that could be included in a gene panel sequencing test for PML risk prediction. Other embodiments could be similarly devised from genes reported in Table 13, or from other tables disclosed herein.


Table 9 contains ‘example’ variants that may be considered as ‘AD’ causes of immunodeficiency (i.e., presence of just 1 of the 2 reported het SNVs in a given patient may be causing immunodeficiency), which may increase the risk for PML. For example, this may be a more likely scenario for het SNVs that are ‘novel’ in the ExAC db (i.e., not found in the general population), and even more likely if such novel SNVs are found in >=2 PML cases (irrespective of the invoked disease model). Examples of this include the following 3 genes:

    • AK2, 2 cases (Table 9)
      • chr1:33476435, C>A, novel in ExAC
      • PML20 and PML33, AFR and EUR, both HIV
    • EPG5, 2 cases (Table 9)
      • chr18: 43445601, T>G, novel in ExAC
      • PML25 and PML27, both EUR, both HIV
    • TNFRSF11A, 9 cases (Table 7)
      • chr18: 60052034, A>C, novel in ExAC
      • see Table 7 for case IDs, 2 AFR and 7 EUR, all HIV


It can be appreciated by those skilled in the art that immunodeficiency genes presently known to cause AR disease may potentially cause AD disease. Numerous examples have been reported in the literature, including several of the genes listed in Table 6 (e.g., Disease_model is indicated as AD_AR for 32 genes, such as ADAR and TICAM1).


Example 15—Exemplary 96-Gene Panel PML Risk Prediction Tests

Table 19 contains an exemplary 96-gene panel based on genes that were found to have at least one PML case count from Tables 7 and 8. The “Genes” and “Case_level_solutions” columns showed genes and total number of PML cases (with at least one ‘case level’ solution) reported in Tables 7 and 8. In addition, the top 7 genes (CHD7, IFIH1, IGLL1, MAVS, PLCG2, SHARPIN, TCIRG1) from Table 14 with SNVs based on ‘PML_ALL_FET’ values <0.05 (column 0) were also included in Table 19. Among these 7 genes, 3 genes (IGLL1, MAVS, SHARPIN) with SNVs were based on ‘PML_ALL_FET’ values <0.05 (column 0) from Table 15.









TABLE 19







exemplary 96-gene panel










Genes
Case_level_solutions













AP3B1
5



APOL1
1



ASH1L
1



ATM
1



ATR
3



BLM
1



CARD11
3



CDKN1B
1



CHD7
4



CLCN7
1



DCLRE1C
3



DDX58
1



DOCK8
8



EGR1
1



EPG5
3



ETF1
1



FPR2
1



GATA2
2



GFI1
4



HIVEP1
1



HIVEP2
2



HTR2A
1



IDO2
1



IFIH1
3



IFNGR2
1



IFNLR1
1



IGLL1
0



IKBKB
1



IL17F
1



IL1B
1



IL21R
1



IRAK4
2



ITSN2
2



JUN
2



KAT6B
1



KCTD7
1



LIG4
1



LRBA
1



MALL
1



MAPK3
2



MAVS
0



MCEE
1



MKL1
1



MYD88
1



NBN
1



NFKB1
3



NOD2
6



NRIP1
1



PIAS1
1



PIAS2
1



PIK3CD
4



PIK3CD-AS1
1



PIK3R1
1



PKHD1
3



PLCG2
5



PNPT1
1



POLA1
1



POLE
1



PRF1
1



PRKCB
1



PRKCD
1



PRKCH
1



PRKDC
4



PSTPIP1
1



PTEN
1



PTPRC
2



RABGEF1
1



RAD51
1



RAG1
4



RAG2
2



RIPK1
1



RIPK3
2



RNF168
2



RTEL1
2



SHARPIN
1



SKIV2L
1



SMAD4
1



STIM1
2



STIM2
1



STXBP2
3



TAP2
1



TBK1
2



TCIRG1
1



TICAM1
2



TLR3
2



TLR4
1



TNFRSF11A
10



TNFRSF13B
1



TNFRSF8
1



TP53
1



TRAF3
1



TRAFD1
1



TRPM2
1



VPS45
1



WEE1
2



ZAP70
3



TOTAL (96 genes)
172



Non-redundant cases
67



Dx yield for PML cohort (n = 70)
95.7%









The non-redundant number of PML cases and diagnostic yield are listed in the last 2 rows of Table 19. Specifically, a test including the 96 genes had a diagnostic yield of 95.7% based on the genetic findings from the 70 PML cases used in the present study.


Example 16—Exemplary 39-Gene Panel PML Risk Prediction Tests

Table 20 contains an exemplary 39-gene panel based on genes that were found to have multiple PML case count from Tables 7 and 8. The “Genes” and “Case_Level_solutions” columns showed genes and total number of PML cases (with at least two ‘case level’ solutions) reported in Tables 7 and 8. In addition, the top 7 genes (CHD7, IFIH1, IGLL1, MAVS, PLCG2, SHARPIN, TCIRG1) from Table 14 with SNVs based on ‘PML_ALL_FET’ values <0.05 (column 0) were also included in Table 20. Among these 7 genes, 3 genes (IGLL1, MAVS, SHARPIN) with SNVs were based on ‘PML_ALL_FET’ values <0.05 (column 0) from Table 15.









TABLE 20







exemplary 39-gene panel










Genes
Case_level_solutions













AP3B1
5



ATR
3



CARD11
3



CHD7
4



DCLRE1C
3



DOCK8
8



EPG5
3



GATA2
2



GFI1
4



HIVEP2
2



IFIH1
3



IGLL1
0



IRAK4
2



ITSN2
2



JUN
2



MAPK3
2



MAVS
0



NFKB1
3



NOD2
6



PIK3CD
4



PKHD1
3



PLCG2
5



PRKDC
4



PTPRC
2



RAG1
4



RAG2
2



RIPK3
2



RNF168
2



RTEL1
2



SHARPIN
1



STIM1
2



STXBP2
3



TBK1
2



TCIRG1
1



TICAM1
2



TLR3
2



TNFRSF11A
10



WEE1
2



ZAP70
3



TOTAL (39 genes)
115



Non-redundant cases
57



Dx yield for PML cohort (n = 70)
81.4%









The non-redundant number of PML cases and diagnostic yield are listed in the last 2 rows of Table 20. Specifically, a test including the 39 genes had a diagnostic yield of 81.4% based on the genetic findings from the 70 PML cases used in the present study.


Example 17—Exemplary 23-Gene Panel PML Risk Prediction Tests

Table 21 contains an exemplary 23-gene panel based on genes that were found to have multiple PML case count from Tables 7 and 8. The “Genes” and “Case_Level_solutions” columns showed genes and total number of PML cases (with at least three ‘case level’ solutions) reported in Tables 7 and 8. In addition, the top 7 genes (CHD7, IFIH1, IGLL1, MAVS, PLCG2, SHARPIN, TCIRG1) from Table 14 with SNVs based on ‘PML_ALL_FET’ values <0.05 (column 0) were also included in Table 21. Among these 7 genes, 3 genes (IGLL1, MAVS, SHARPIN) with SNVs were based on ‘PML_ALL_FET’ values <0.05 (column 0) from Table 15.









TABLE 21







exemplary 23-gene panel










Genes
Case_level_solutions













AP3B1
5



ATR
3



CARD11
3



CHD7
4



DCLRE1C
3



DOCK8
8



EPG5
3



GFI1
4



IFIH1
3



IGLL1
0



MAVS
0



NFKB1
3



NOD2
6



PIK3CD
4



PKHD1
3



PLCG2
5



PRKDC
4



RAG1
4



SHARPIN
1



STXBP2
3



TCIRG1
1



TNFRSF11A
10



ZAP70
3



TOTAL (23 genes)
83



Non-redundant cases
50



Dx yield for PML cohort (n = 70)
71.4%









The non-redundant number of PML cases and diagnostic yield are listed in the last 2 rows of Table 21. Specifically, a test including the 23 genes had a diagnostic yield of 71.4% based on the genetic findings from the 70 PML cases used in the present study.


Example 18—Exemplary 15-Gene Panel PML Risk Prediction Tests

Table 22 contains an exemplary 15-gene panel based on genes that were found to have multiple PML case count from Tables 7 and 8. The “Genes” and “Case_Level_solutions” columns showed genes and total number of PML cases (with at least four ‘case level’ solutions) reported in Tables 7 and 8. In addition, the top 7 genes (CHD7, IFIH1, IGLL1, MAVS, PLCG2, SHARPIN, TCIRG1) from Table 14 with SNVs based on ‘PML_ALL_FET’ values <0.05 (column 0) were also included in Table 22. Among these 7 genes, 3 genes (IGLL1, MAVS, SHARPIN) with SNVs were based on ‘PML_ALL_FET’ values <0.05 (column 0) from Table 15.









TABLE 22







exemplary 15-gene panel










Genes
Case_level_solutions













AP3B1
5



CHD7
4



DOCK8
8



GFI1
4



IFIH1
3



IGLL1
0



MAVS
0



NOD2
6



PIK3CD
4



PLCG2
5



PRKDC
4



RAG1
4



SHARPIN
1



TCIRG1
1



TNFRSF11A
10



TOTAL (15 genes)
59



Non-redundant cases
39



Dx yield for PML cohort (n = 70)
55.7%









The non-redundant number of PML cases and diagnostic yield are listed in the last 2 rows of Table 22. Specifically, a test including the 15 genes had a diagnostic yield of 55.7% based on the genetic findings from the 70 PML cases used in the present study.


Example 19— Exemplary 11-Gene Panel PML Risk Prediction Tests

Table 23 contains an exemplary 11-gene panel based on genes that were found to have multiple PML case count from Tables 7 and 8. The “Genes” and “Case_Level_solutions” columns showed genes and total number of PML cases (with at least five ‘case level’ solutions) reported in Tables 7 and 8. In addition, the top 7 genes (CHD7, IFIH1, IGLL1, MAVS, PLCG2, SHARPIN, TCIRG1) from Table 14 with SNVs based on ‘PML_ALL_FET’ values <0.05 (column 0) were also included in Table 23. Among these 7 genes, 3 genes (IGLL1, MAVS, SHARPIN) with SNVs were based on ‘PML_ALL_FET’ values <0.05 (column 0) from Table 15.









TABLE 23







exemplary 11-gene panel










Genes
Case_level_solutions













AP3B1
5



CHD7
4



DOCK8
8



IFIH1
3



IGLL1
0



MAVS
0



NOD2
6



PLCG2
5



SHARPIN
1



TCIRG1
1



TNFRSF11A
10



TOTAL (11 genes)
43



Non-redundant cases
33



Dx yield for PML cohort (n = 70)
47.1%









The non-redundant number of PML cases and diagnostic yield are listed in the last 2 rows of Table 23. Specifically, a test including the 11 genes had a diagnostic yield of 47.1% based on the genetic findings from the 70 PML cases used in the present study.


Example 20—Exemplary 10-Gene Panel PML Risk Prediction Tests

Table 24 contains an exemplary 10-SNV panel based on top 7 SNVs in Table 14 and 3 SNVs from Table 15 (based on overlapping genes between 14 and 15: IGLL1, MAVS, SHARPIN). Specifically, Using the top 10 SNVs (7 from Table 14, along with 3 from Table 15, residing in genes already selected from Table 14), an additive count (column “Case total additive (non-redundant)”) was performed to determine how many PML cases had at least one of the variants when these were considered in order (e.g., column “Order (FET)”: ‘1’, first, followed by ‘1’+‘2’, followed by ‘1’+‘2’+‘3’, etc). Since some individuals harbor more than one variant, the additive count is not equal to the simple sum of PML case numbers for each variant (column “Case total per SNV”). All genome coordinates are based on hg19 build.


An additive count was performed for ExAC subjects (column “ExAC subjects total additive (redundant)”), as follows: i) The average cohort size for ExAC for all variants was calculated; ii) Each total subject count (all ethnicities) was normalized to this average cohort size. The ExAC additive count represents a simple addition: labeled as “redundant” in column “ExAC subjects total additive (redundant)”, because information regarding the possible presence of multiple variants in the same individual is not available; iii) Odds Ratios (ORs) and Fisher's Exact test (FET) values were calculated (columns “PML ALL OR additive” and “PML ALL FET additive”).









TABLE 24







exemplary 10-gene panel

























ExAC









Case
Case total

subjects
PML
PML







total
additive
Dx yield
total
ALL
ALL


Order
Table

Variant

per
(non-
(non-
additive
OR
FET


(FET)1
source
Gene
(hg19)
Genotype
SNV
redundant)2
redundant)
(redundant)3
additive
additive




















1
14
PLCG2
chr16: 81942175,
het
7
7
10%
730
6.50
2.00E−04





A > G









2
14
IFIH1
chr2: 163136505,
het
7
13
19%
1,473
6.49
6.37E−07





C > G









3
14
TCIRG1
chr11: 67818269,
het
4
16
23%
1,830
6.73
2.94E−08





G > A









4
14
IGLL1
chr22: 23917192,
het
8
22
31%
3,388
5.42
9.41E−09





G > T









5
14
MAVS
chr20: 3846397,
hom
8
26
37%
4,947
4.60
2.13E−08





C > T









6
14
SHARPIN
chr8: 145154222,
het
12
33
47%
8,064
3.91
5.10E−08





G > A









7
14
CHD7
chr8: 61654298,
het
5
36
51%
9,292
3.89
3.26E−08





T > A









8
15
SHARPIN
chr8: 145154824,
het
3
37
53%
9,294
4.12
8.10E−09





A > C









9
15
IGLL1
chr22: 23915745,
het
3
38
54%
9,394
4.30
2.59E−09





G > A









10
15
MAVS
chr20: 3843027,
hom
6
38
54%
10,393
3.77
5.26E−08





C > A






1SNV order based on lowest FET value reported in Tables 14 and 15 for combined ethnicities




2PML case total = 70




3ExAC subject total = 43,419 (average for the 10 SNVs)







It can be appreciated by those skilled in the art that the above gene panels were selected based on the present genetic findings in 70 PML cases. Furthermore, a gene not presently selected for any of these exemplary gene panels may be added to the gene panel. For example, genes in which only 1 PML case was found to have variants fulfilling the criteria may be added to the gene panel if genetic validation in additional PML cases shows a ‘n=1 case’ gene is impacted by more than 1 PML case when the data are examined for a new set of PML cases. In some cases, additional genes (e.g., PML-linked genes such as DOCKS, BAG3, STAT1) may be added to the gene panel.


While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims
  • 1. A method of treating a condition in a subject in need of belimumab comprising: administering a therapeutically effective amount of belimumab to the subject, wherein the subject has a decreased risk of progressive multifocal leukoencephalopathy (PML) due to an infection of the brain by John Cunningham virus (JCV), wherein the subject's decreased risk is associated with the absence of one or more genetic variations in the subject, wherein the subject has been tested for a presence of the one or more genetic variations with a genetic assay and has been identified as not having the one or more genetic variations; wherein the one or more genetic variations comprise a single nucleotide variation (SNV) in a syntaxin binding protein 2 (STXBP2) gene, wherein the SNV in a STXBP2 gene comprises chr19:7712287 G>C; wherein chromosome positions of the one or more genetic variations are defined with respect to UCSC hg19.
  • 2. The method of claim 1, wherein the condition is lupus.
  • 3. The method of claim 2, wherein the condition is systemic lupus erythematosus.
  • 4. The method of claim 1, wherein the subject has been identified as not having one or more other genetic variations that disrupt or modulate a corresponding gene according to Tables 1, 3, 6-10, 28A, 29, 31, 34-36, 47 and 48.
  • 5. The method of claim 1, wherein the subject has been identified as not having one or more other genetic variations that disrupt or modulate a corresponding gene according to Tables 19-24, 40 and 42.
  • 6. The method of claim 1, wherein the subject has been tested with a JCV-antibody test, a CD62L test, or a CSF IgM oligoclonal bands test.
  • 7. The method of claim 1, wherein the method further comprises testing the subject for the presence of the one or more genetic variations with the genetic assay prior to the administering.
  • 8. The method of claim 7, wherein the genetic assay comprises microarray analysis, PCR, sequencing, nucleic acid hybridization, or any combination thereof.
  • 9. The method of claim 7, wherein prior to testing the subject for the presence of the one or more genetic variations with the genetic assay the method further comprises obtaining biological samples from subjects with PML and (a) confirming each biological sample is not a duplicate of any other biological sample based on nucleic acid information of the biological samples or (b) determining a sex genotype for each biological sample based on nucleic acid information of the biological samples, and confirming the sex genotype of each biological sample is the same as a sex phenotype of the subject with PML from which the biological sample was obtained.
CROSS-REFERENCE

This application is a divisional of U.S. application Ser. No. 17/141,885, filed Jan. 5, 2021, which is a divisional of U.S. application Ser. No. 16/245,849, filed Jan. 11, 2019, now U.S. Pat. No. 10,941,448, issued Mar. 9, 2021, which is a continuation of U.S. application Ser. No. 15/639,591 filed Jun. 30, 2017, now U.S. Pat. No. 10,240,205, issued Mar. 26, 2019, which claims the benefit of U.S. Provisional Application No. 62/524,324, filed Jun. 23, 2017, and U.S. Provisional Application No. 62/454,676, filed Feb. 3, 2017, both of which are incorporated herein by reference in their entireties.

US Referenced Citations (155)
Number Name Date Kind
696589 Pleck Apr 1902 A
3625214 Higuchi Dec 1971 A
4683195 Mullis et al. Jul 1987 A
4789734 Pierschbacher Dec 1988 A
4897268 Tice et al. Jan 1990 A
4906474 Langer et al. Mar 1990 A
4925673 Steiner et al. May 1990 A
4992445 Lawter et al. Feb 1991 A
5001139 Lawter et al. Mar 1991 A
5023252 Hseih Jun 1991 A
5075109 Tice et al. Dec 1991 A
5143854 Pirrung et al. Sep 1992 A
5190029 Byron et al. Mar 1993 A
5223409 Ladner et al. Jun 1993 A
5272071 Chappel Dec 1993 A
5288514 Ellman Feb 1994 A
5376359 Johnson Dec 1994 A
5384261 Winkler et al. Jan 1995 A
5424186 Fodor et al. Jun 1995 A
5445934 Fodor et al. Aug 1995 A
5451683 Barrett et al. Sep 1995 A
5510270 Fodor et al. Apr 1996 A
5527681 Holmes Jun 1996 A
5665549 Pinkel et al. Sep 1997 A
5700637 Southern Dec 1997 A
5744305 Fodor et al. Apr 1998 A
5776434 Purewal et al. Jul 1998 A
5811128 Tice et al. Sep 1998 A
5814344 Tice et al. Sep 1998 A
5820883 Tice et al. Oct 1998 A
5837832 Chee et al. Nov 1998 A
5853763 Tice et al. Dec 1998 A
5858659 Sapolsky et al. Jan 1999 A
5928647 Rock Jul 1999 A
5942252 Tice et al. Aug 1999 A
5945334 Besemer et al. Aug 1999 A
6040138 Lockhart et al. Mar 2000 A
6054270 Southern Apr 2000 A
6146834 Schaad et al. Nov 2000 A
6207392 Weiss et al. Mar 2001 B1
6210878 Pinkel et al. Apr 2001 B1
6251607 Tsen et al. Jun 2001 B1
6300063 Lipshutz et al. Oct 2001 B1
6423499 Song et al. Jul 2002 B1
6429027 Chee et al. Aug 2002 B1
6733977 Besemer et al. May 2004 B2
6858394 Chee et al. Feb 2005 B1
6858412 Willis et al. Feb 2005 B2
6892141 Nakae et al. May 2005 B1
6916621 Shah Jul 2005 B2
6951761 Star et al. Oct 2005 B2
6969589 Patil et al. Nov 2005 B2
6977148 Dean et al. Dec 2005 B2
7011949 Amorese et al. Mar 2006 B2
7014997 Knoll et al. Mar 2006 B2
7030231 Craik et al. Apr 2006 B1
7034144 Van Dongen et al. Apr 2006 B2
7238484 Pinkel et al. Jul 2007 B2
7364858 Barany et al. Apr 2008 B2
7424368 Huang et al. Sep 2008 B2
7702468 Chinitz et al. Apr 2010 B2
7910353 Shaffer et al. Mar 2011 B2
7957913 Chinitz et al. Jun 2011 B2
7985913 Machell Jul 2011 B2
7998744 Stevenson et al. Aug 2011 B2
8367417 Stevenson et al. Feb 2013 B2
8655599 Chinitz et al. Feb 2014 B2
8862410 Hatchwell et al. Oct 2014 B2
10059997 Hatchwell et al. Aug 2018 B2
10240205 Hatchwell et al. Mar 2019 B2
10544463 Hatchwell et al. Jan 2020 B2
10563264 Hatchwell Feb 2020 B2
10597721 Hatchwell et al. Mar 2020 B2
10724096 Hatchwell et al. Jul 2020 B2
10941448 Hatchwell et al. Mar 2021 B1
10961585 Hatchwell et al. Mar 2021 B2
20020012921 Stanton Jan 2002 A1
20020012930 Rothberg et al. Jan 2002 A1
20020112930 Arita et al. Aug 2002 A1
20030023070 Ni et al. Jan 2003 A1
20030049663 Wigler et al. Mar 2003 A1
20030068629 Rothberg et al. Apr 2003 A1
20030082606 Lebo et al. May 2003 A1
20030100102 Rothberg et al. May 2003 A1
20030148344 Rothberg et al. Aug 2003 A1
20030207295 Gunderson et al. Nov 2003 A1
20030215821 Gunderson et al. Nov 2003 A1
20040018491 Gunderson et al. Jan 2004 A1
20040137473 Wigler et al. Jul 2004 A1
20040157243 Huang et al. Aug 2004 A1
20040197774 Wigler Oct 2004 A1
20040248161 Rothberg et al. Dec 2004 A1
20050032095 Wigler et al. Feb 2005 A1
20050037393 Gunderson et al. Feb 2005 A1
20050037414 Lee et al. Feb 2005 A1
20050079510 Berka et al. Apr 2005 A1
20050100893 Gunderson et al. May 2005 A1
20050100932 Lapidus et al. May 2005 A1
20050112595 Zhao et al. May 2005 A1
20050124022 Srinivasan et al. Jun 2005 A1
20050196799 Wigler et al. Sep 2005 A1
20050233339 Barrett et al. Oct 2005 A1
20050266444 Wigler et al. Dec 2005 A1
20050282196 Costa Dec 2005 A1
20060012784 Ulmer Jan 2006 A1
20060012793 Harris Jan 2006 A1
20060024678 Buzby Feb 2006 A1
20060024711 Lapidus et al. Feb 2006 A1
20060063168 Albertson et al. Mar 2006 A1
20060078909 Srinivasan et al. Apr 2006 A1
20060134674 Huang et al. Jun 2006 A1
20070141577 Moore Jun 2007 A1
20070207141 Lieberburg Sep 2007 A1
20070207481 Wigler et al. Sep 2007 A1
20070259351 Chinitz et al. Nov 2007 A1
20080131887 Stephan et al. Jun 2008 A1
20080305967 Ward et al. Dec 2008 A1
20090026082 Rothberg et al. Jan 2009 A1
20090098547 Ghosh Apr 2009 A1
20090127589 Rothberg et al. May 2009 A1
20090170712 Beatty et al. Jul 2009 A1
20090304653 Messier Dec 2009 A1
20100003685 Aasly et al. Jan 2010 A1
20100028931 Eggan et al. Feb 2010 A1
20100035252 Rothberg et al. Feb 2010 A1
20100120046 Brennan et al. May 2010 A1
20100137143 Rothberg et al. Jun 2010 A1
20100167286 Reijo Pera et al. Jul 2010 A1
20100188073 Rothberg et al. Jul 2010 A1
20100197507 Rothberg et al. Aug 2010 A1
20100227768 Wigler et al. Sep 2010 A1
20100248236 Chinitz et al. Sep 2010 A1
20110021366 Chinitz et al. Jan 2011 A1
20110111014 Langston May 2011 A1
20110130337 Eriksson et al. Jun 2011 A1
20110264376 Chinitz et al. Oct 2011 A1
20110311512 Hakonarson et al. Dec 2011 A1
20120059594 Hatchwell et al. Mar 2012 A1
20120100995 Scherer et al. Apr 2012 A1
20130247249 Singh et al. Sep 2013 A1
20130305410 Bent et al. Nov 2013 A1
20130316911 Scherer Nov 2013 A1
20140088882 Chinitz et al. Mar 2014 A1
20140155271 Hatchwell et al. Jun 2014 A1
20140161721 Hatchwell et al. Jun 2014 A1
20140162894 Hatchwell et al. Jun 2014 A1
20140162933 Hatchwell et al. Jun 2014 A1
20140208449 Malek Jul 2014 A1
20150051086 Hatchwell et al. Feb 2015 A1
20150132295 Hatchwell et al. May 2015 A1
20160019336 Chinitz et al. Jan 2016 A1
20170016919 Plavina et al. Jan 2017 A1
20190071726 Hatchwell et al. Mar 2019 A1
20210371928 Hatchwell et al. Dec 2021 A1
20210381052 Hatchwell et al. Dec 2021 A1
Foreign Referenced Citations (60)
Number Date Country
1733937 Feb 2006 CN
101148684 Mar 2008 CN
101403008 Apr 2009 CN
103436606 Dec 2013 CN
0373203 Jun 1990 EP
0373203 Aug 1994 EP
0619321 Oct 1994 EP
20090080105 Jul 2009 KR
20110114664 Oct 2011 KR
WO-9002809 Mar 1990 WO
WO-9106667 May 1991 WO
WO-9117271 Nov 1991 WO
WO-9201047 Jan 1992 WO
WO-9209690 Jun 1992 WO
WO-9210092 Jun 1992 WO
WO-9215679 Sep 1992 WO
WO-9218619 Oct 1992 WO
WO-9220791 Nov 1992 WO
WO-9209690 Dec 1992 WO
WO-9301288 Jan 1993 WO
WO-9322684 Nov 1993 WO
WO-9511995 May 1995 WO
WO-9820019 May 1998 WO
WO-02099129 Dec 2002 WO
WO-03048318 Jun 2003 WO
WO-2004018633 Mar 2004 WO
WO-2004044225 May 2004 WO
WO-2004075010 Sep 2004 WO
WO-2005042763 May 2005 WO
WO-2005068664 Jul 2005 WO
WO-2005108997 Nov 2005 WO
WO-2004044225 Apr 2006 WO
WO-2006050475 May 2006 WO
WO-2006091254 Aug 2006 WO
WO-2006116873 Nov 2006 WO
WO-2007070640 Jun 2007 WO
WO-2007070640 Aug 2007 WO
WO-2007129000 Nov 2007 WO
WO-2007131135 Nov 2007 WO
WO-2008016374 Feb 2008 WO
WO-2007129000 Mar 2008 WO
WO-2007131135 Nov 2008 WO
WO-2009038684 Mar 2009 WO
WO-2009043178 Apr 2009 WO
WO-2009073764 Jun 2009 WO
WO-2010036353 Apr 2010 WO
WO-2010056897 May 2010 WO
WO-2010124101 Oct 2010 WO
WO-2011012672 Feb 2011 WO
WO-2011035012 Mar 2011 WO
WO-2011112961 Sep 2011 WO
WO-2012023519 Feb 2012 WO
WO-2012027491 Mar 2012 WO
WO-2012047234 Apr 2012 WO
WO-2013067451 May 2013 WO
WO-2013071119 May 2013 WO
WO-2014043519 Mar 2014 WO
WO-2015131078 Sep 2015 WO
WO-2018158632 Sep 2018 WO
WO-2020033700 Feb 2020 WO
Non-Patent Literature Citations (340)
Entry
Hatchwell et al. Progressive multifocal leukoencephalopathy genetic risk variants for pharmacovigilance of immunosuppressant therapies. Front. Neurol. 13:1016377. doi: 10.3389/fneur. 2022.1016377.
Abravaya, et al. Detection of point mutations with a modified ligase chain reaction (Gap-LCR). Nucleic Acids Research. 1995;23(4):675-682.
Agami, R. RNAi and related mechanisms and their potential use for therapy. Curr Opin Chem Biol. Dec. 2002;6(6):829-34.
Agarwal et al., Novelty in the target landscape of the pharmaceutical. Nat. Rev. Drug Discovery 12(8):575-6 (2013).
Aitman, et al. Copy number polymorphism in Fcgr3 predisposes to glomerulonephritis in rats and humans. Nature. Feb. 16, 2006;439(7078):851-5.
Albertson, et al. Profiling breast cancer by array CGH. Breast Cancer Res Treat. Apr. 2003;78(3):289-98.
Alexander Zimprich, et al., A mutation in, encoding a subunit of the retromer complex, causes late-onset parkinson disease, American journal of human genetics, American society of human genetics. Jun. 2011; 89(1):168-175.
Altschul, et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. Sep. 1, 1997;25(17):3389-402.
Amarzguioui, et al. Approaches for chemically synthesized siRNA and vector-mediated RNAi. FEBS Lett. Oct. 31, 2005;579(26):5974-81. Epub Sep. 20, 2005.
Ansel, Howard C, et al. Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia, PA: Lippincott-Williams & Wilkins, 1999. Print.
Antoniol, et al., Immunological markers for PML prediction in MS patients treated with natalizumab. Frontiers in Immunology. Jan. 2015; 5(668): pp. 1-9.
Arakawa, et al. Advances in characterization of neuroprotective peptide, humanin. Curr Med Chem. 2011;18(36):5554-63.
Ausubel (Ed.), Current Protocols in Molecular Biology (2007 John Wiley & Sons, NY).
Ausubel, et al. Current Protocols in Molecular Biology. John Wiley & Sons, New York, 1999.
Bailey, et al. Analysis of Segmental Duplications and Genome Assembly in the Mouse. Genome Res. 2004; 14:789-801.
Bakkaloglu, et al. Molecular cytogenetic analysis and resequencing of contactin associated protein-like 2 in autism spectrum disorders. Am J Hum Genet. Jan. 2008;82(1):165-73.
Bangham et al. Diffusion of univalent ions across the lamellae of swollen phospholipids. J. Mol. Biol. 1965;13:238-252.
Bedell, et al. In vivo genome editing using a high-efficiency TALEN. Nature. 491.7422 (2012):114-118.
Benjamini e al. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society. Series B (Methodological). pp. 289-300 (1995).
Bennett, C. Efficiency of antisense oligonucleotide drug discovery. Antisense Nucleic Acid Drug Dev. Jun. 2002;12(3):215-24.
Berkel, et al. Mutations in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental retardation. Nat Genet. Jun. 2010;42(6):489-91. Epub May 16, 2010.
Bernard, et al. Sequence of the murine and human cellular myc oncogenes and two modes of myc transcription resulting from chromosome translocation in B lymphoid tumours. EMBO J. 1983;2(12):2375-83.
Bernstein, et al. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature. Jan. 18, 2001;409(6818):363-6.
Betancur, et al. The emerging role of synaptic cell-adhesion pathways in the pathogenesis of autism spectrum disorders. Trends Neurosci. Jul. 2009;32(7):402-12. doi: 10.1016/j.tins.2009.04.003. Epub Jun. 21, 2009.
Bier, et al. DNA microarrays. Adv Biochem Eng Biotechnol. 2008;109:433-53.
Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. Mar. 2001;69(3):89-95.
Bochukova, et al. Large, rare chromosomal deletions associated with severe early-onset obesity. Nature. Feb. 4, 2010;463(7281):666-70. Epub Dec. 6, 2009.
Bodmer, et al. Common and rare variants in multifactorial susceptibility to common diseases. Nat Genet. Jun. 2008;40(6):695-701.
Bodzioch, et al. Evidence for potential functionality of nuclearly-encoded humanin isoforms. Genomics. Oct. 2009;94(4):247-56. Epub May 27, 2009.
Bosher, et al. RNA interference: genetic wand and genetic watchdog. Nat Cell Biol. Feb. 2000;2(2):E31-6.
Bowen, et al., HIV-associated opportunistic CNS infections: pathophysiology, diagnosis and treatment.Nat Rev Neurol. Oct. 27, 2016;12(11):662-674. doi: 10.1038/nrneurol.2016.149.
Bremer, et al. Copy number variation characteristics in subpopulations of patients with autism spectrum disorders. Am J Med Genet B Neuropsychiatr Genet. Mar. 2011;156(2):115-24. doi: 10.1002/ajmg.b.31142. Epub Dec. 8, 2010.
Brummelkamp, et al. A system for stable expression of short interfering RNAs in mammalian cells. Science. Apr. 19, 2002;296(5567):550-3. Epub Mar. 21, 2002.
Bult, et al. The Mouse genome Database (MGD): mouse biology and model systems. Nucleic Acids Research. 2008; 36 Database Issue: D724-D728. doi:10.1093/nar/gkm961.
Bych, et al., The iron-sulphur protein Ind1 is required for effective complex I assembly. The EMBO Journal (2008) 27, 1736-174.
Calabrese, et al., Sorting out the risks in progressive multifocal leukoencephalopathy. Nat Rev Rheumatol. Feb. 2015;11(2):119-23. doi: 10.1038/nrrheum.2014.167. Epub Oct. 14, 2014.
Calabrese, et al., Sorting out the risks in progressive multifocal leukoencephalopathy. Nat. Rev. Rheumatol. advance online publication Oct. 14, 2014; pp. 1-5.
Calvo, et al. High-throughput, pooled sequencing identifies mutations in NUBPL and FOXRED1 in human complex I deficiency. Nat Genet. Oct. 2010;42(10):851-8. Epub Sep. 5, 2010.
Carles Vilario-Guell, et al., Mutations in Parkinson disease, American journal of human genetics, american society of human genetics. Jun. 2011; 89(1):162-167.
Chahin, et al., A risk classification for immunosuppressive treatment-associated progressive multifocal leukoencephalopathy. J. Neurovirol. (2015) 21:623-631 DOI 10.1007/s13365-014-0303-1.
Chan, et al., Identification of key residues essential for the structural fold and receptor selectivity within the A-chain of human gene-2 (H2) relaxin. The Journal of Biological Chemistry vol. 287, No. 49, pp. 41152-41164, Nov. 30, 2012.
Chavanpatil et al. Novel sustained release, swellable and bioadhesive gastroretentive drug delivery system for olfoxacin. International Journal of Pharmaceutics. 2006;316(1-2):86-92.
Chen, et al., Correlation between SMN2 copies and the phenotype of spinal muscular atrophy. Chin J Neurol, Nov. 30, 2005; 38(11):673-676.
Chen, et al., Identification of small molecule agonists of human relaxin family receptor 1 (RXFP1) by utilizing a homogenous cell-based cAMP assay.
Chen, et al. The evolution of gene regulation by transcription factors and microRNAs. Nat Rev Genet. Feb. 2007;8(2):93-103.
Chen, H. Clinical development of antisense oligonucleotides as anti-cancer therapeutics. Methods Mol Med. 2003;75:621-36.
Chi, et al. Genomewide view of gene silencing by small interfering RNAs. Proc Natl Acad Sci U S A. May 27, 2003;100(11):6343-6. Epub May 2, 2003.
Ching, et al., Integrated analysis of copy number and loss of heterozygosity in primary breast carcinomas using high-density SNP array. International journal of oncology, 2011; 39:621-633.
Chinn, et al., Severe combined immunodeficiency disorders. Immunol Allergy Clin N Am 35 (2015) 671-694.
CNV: 14q23.3 summary output from https://gene.sfari.org/database/cnv/14q23.3 Nov. 30, 2017, pp. 1-3. (year: 2017).
Cohen, Jonathan C. “Emerging LDL therapies: Using human genetics to discover new therapeutic targets for plasma lipids.” Journal of clinical lipidology vol. 7,3 Suppl (2013): S1-5. doi:10.1016/j.jacl.2013.03.005.
Colin et al.,Drug-induced progressive multifocal leukoencephalopathy: a case/noncase study in the French pharmacovigilance database. Fundam Clin Pharmacol. Oct. 13, 2016: pp. 1-8.
Conrad, et al. Origins and functional impact of copy number variation in the human genome. Nature. Apr. 1, 2010;464(7289):704-12. Epub Oct. 7, 2009.
Copy Number Variants summary for 12q23.3-q24.13 from gene.sfari.org/database/cnv/, two pages printed on Dec. 2, 2017. (Year:2017).
Corti, et al. What Genetics tells us about the causes and mechanisms of parkinson's disease. Physiological reviews.Oct. 2011; 91(4): 1161-1218.
Coskun, et al., A Mitochondrial Etiology of Alzheimer and Parkinson Disease. Biochim Biophys Acta. May 2012 ; 1820(5): 553-564. doi:10.1016/j.bbagen.2011.08.008.
Crespi, et al. Association testing of copy number variants in schizophrenia and autism spectrum disorders. J Neurodev Disord. May 30, 2012;4(1):15. doi: 10.1186/1866-1955-4-15.
Cronin, et al. Analysis of genome-wide copy number variation in Irish and Dutch ALS populations. Hum Mol Genet. Nov. 1, 2008;17(21):3392-8. Epub Aug. 7, 2008.
D'Amico, et al., Treatment-related progressive multifocal leukoencephalopathy in multiple sclerosis: A comprehensive review of current evidence and future needs. Drug Saf. 2016;39:1163-1174.
Daruwala, et al. A versatile statistical analysis algorithm to detect genome copy number variation. Proc Natl Acad Sci U S A. Nov. 16, 2004;101(46):16292-7. Epub Nov. 8, 2004.
Day-Williams, et al., Whole Genome Sequencing Reveals a Chromosome 9p Deletion Causing DOCK8 Deficiency in an Adult Diagnosed with Hyper IgE Syndrome Who Developed Progressive Multifocal Leukoencephalopathy. J Clin Immunol (2015) 35:92-96; DOI 10.1007/s10875-014-0114-4.
De Krom, et al. A common variant in DRD3 receptor is associated with autism spectrum disorder. Biol Psychiatry. Apr. 1, 2009;65(7):625-30. doi: 10.1016/j.biopsych.2008.09.035. Epub Dec. 5, 2008.
Desmet, et al., Human Splicing Finder: an online bioinformatics tool to predict splicing signals. Nucleic Acids Research, 2009, vol. 37, No. 9 e67.
Dias, et al. Antisense oligonucleotides: basic concepts and mechanisms. Mol Cancer Ther. Mar. 2002;1(5):347-55.
Dibbens, et al. Familial and sporadic 15q13.3 microdeletions in Idiopathic Generalized Epilepsy: Precedent for Disorders with Complex Inheritance. Hum Mol Genet. Jul. 10, 2009. [Epub ahead of print].
Dijkhuizen, et al. FISH and array-CGH analysis of a complex chromosome 3 aberration suggests that loss of CNTN4 and CRBN contributes to mental retardation in 3pter deletions. Am J Med Genet A. Nov. 15, 2006;140(22):2482-7.
Durandy, et al., Primary antibody deficiencies.Nature Reviews: Immunology. Jul. 2013; 13: pp. 519-533.
Durandy, et al., Supplementary Information Table 1. Nature Reviews: Immunology. Jul. 2013; 1 page.
Durandy, et al., Supplementary Information Table 2. Nature Reviews: Immunology. Jul. 2013; 1 page.
Elbashir, et al. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev. Jan. 15, 2001;15(2):188-200.
Encode project consortium, et al. An integrated encyclopedia of DNA elements in the human genome. Nature. Sep. 6, 2012;489(7414):57-74. doi: 10.1038/nature11247.
Estivill, et al. Copy number variants and common disorders: filling the gaps and exploring complexity in genome-wide association studies. PLoS Genet. Oct. 2007;3(10):1787-99.
ExAC Browser (BETA) | Exome aggregation consortium. Available at http://exac.broadinstitute.org/. Accessed on Jun. 8, 2017.
Fan, et al. Illumina universal bead arrays. Methods Enzymol. 2006;410:57-73.
Fassbender, A., et al., “Biomarkers of endometriosis”, Fertility and Sterility, 99(4), (Mar. 15, 2013), 1135-1145.
Fassone, et al., Complex I deficiency: clinical features, biochemistry and molecular genetics. J Med Genet 2012;49:578-590. doi:10.1136/jmedgenet-2012-101159.
Fernandez, et al. Disruption of contactin 4 (CNTN4) results in developmental delay and other features of 3p deletion syndrome. Addendum. Am J Hum Genet. Jun. 2008;82(6):1385.
Fernandez, et al. Disruption of contactin 4 (CNTN4) results in developmental delay and other features of 3p deletion syndrome. Am J Hum Genet. Jun. 2004;74(6):1286-93.
Fernandez, et al. Gene Discovery in Developmental Neuropsychiatric Disorders: Clues from Chromosomal Rearrangements. Yale Journal of Biology and Medicine, vol. 78 (2005), pp. 95-130. on p. 103. Abstract.
Fire et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391 (1998): 806-811.
Flannick, et al., Loss-of-function mutations in SLC30A8 protect against type 2 diabetes. Nat Genet. Author manuscript; available in PMC Jun. 10, 2014.
Freeman, et al. Copy number variation: new insights in genome diversity. Genome Res. Aug. 2006;16(8):949-61. Epub Jun. 29, 2006.
Freshney, R. I., Culture of Animal Cells: A Manual of Basic Technique. Wiley-Liss; 5th edition (2005).
Gagne, et al. Natalizumab-related PML 2 weeks after negative anti-jcv antibody assay. Neurology. 2016; 86:484-486.
Gagneux, et al. Genetic differences between humans and great apes. Mol Phylogenet Evol. Jan. 2001;18(1):2-13.
Galfre et al. Antibodies to major histocompatibility antigens produced by hybrid cell lines. Nature 266:550-552 (1977).
Gatto, et al. Genetic controls balancing excitatory and inhibitory synaptogenesis in neurodevelopmental disorder models. Frontiers in Synaptic Neuroscience. Jun. 2010; 2(4):1-19.
Gelb, et al., Diagnostic Criteria for Parkinson Disease. Arch Neurol. 1999;56(1):33-39. doi:10.1001/archneur.56.1.33.
Gelmann, et al. Identification of reciprocal translocation sites within the c-myc oncogene and immunoglobulin mu locus in a Burkitt lymphoma. Nature. Dec. 22, 1983-Jan. 4, 1984;306(5945):799-803.
GeneCards output for ATXN2 gene, from www.genecards.ord, pritned on May 20, 2015, pp. 1-13.
GeneCards output for DIAPH2 gene, from www.genecards.ord, printed on Jun. 11, 2015, pp. 1-11.
Gheuens, et al., Role of human leukocyte antigen class I alleles in progressive multifocal leukoencephalopathy. Journal of NeuroVirology, 2010; 1-7.
Gilling, et al. Breakpoint cloning and haplotype analysis indicate a single origin of the common Inv(10)(p11.2q21.2) mutation among northern Europeans. Am. J. Hum. Genet. 2006;78(5):878-83.
Glessner, et al. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature. May 28, 2009;459(7246):569-73. Epub Apr. 28, 2009.
Gokcumen, et al., Copy number variants (CNVs) in primate species using array-based comparative genomic hybridization. Methods 2009;49:18-25.
Goldstein. Common genetic variation and human traits. N Engl J Med. Apr. 23, 2009;360(17):1696-8. Epub Apr. 15, 2009.
GPHN Gene—GeneCards output. pp. 1-14. Printed on Jul. 2, 2015 from www.genecards.org.
Gregoriadis. Chapter 14: Liposomes. Drug Carriers in Biology and Medicine (57 pgs) (Academic Press, 1979).
Gribble, et al. The complex nature of constitutional de novo apparently balanced translocations in patients presenting with abnormal phenotypes. J. Med. Genet. 2005; 42:8-16.
Griffiths, et al. Human anti-self antibodies with high specificity from phage display libraries. EMBO J. Feb. 1993;12(2):725-34.
Griswold, et al. A de novo 1.5 Mb microdeletion on chromosome 14q23.2-23.3 in a patient with autism and spherocytosis. Autism Res. Jun. 2011;4(3):221-7. doi: 10.1002/aur.186. Epub Feb. 28, 2011.
Grskovic, et al. Induced pluripotent stem cells—opportunities for disease modelling and drug discovery. Nat Rev Drug Discov. Nov. 11, 2011;10(12):915-29. doi: 10.1038/nrd3577.
Guatelli et al. Isothermal, in vitro amplification of nucleic acids by a multienzyme reaction modeled after retroviral replication. PNAS USA 87(5):1874-1878 (1990).
Guerini, Franca R. et al., “Analysis of CCR5, CCR2, SDF1 and RANTES gene polymorphisms in subjects with HIV-related PML and not determined leukoencephalopathy”, 2008, Biomedicine & Pharmacotherapy, vol. 62, No. 1, pp. 26-30.
Guilmatre, et al. Recurrent rearrangements in synaptic and neurodevelopmental genes and shared biologic pathways in schizophrenia, autism, and mental retardation. Arch Gen Psychiatry. Sep. 2009;66(9):947-56. doi: 10.1001/archgenpsychiatry.2009.80.
Harada, et al. Subtelomere specific microarray based comparative genomic hybridisation: a rapid detection system for cryptic rearrangements in idiopathic mental retardation. J. Med. Genet. 2004; 41:130-136.
Hastings PJ, Ira G, Lupski JR, (2009) “A Microhomology-Mediated Break-Induced Replication Model for the Origin of Human Copy Number Variation” PLOS Genetics 5(1): e1000327. https://doi.org/10.1371/journal.pgen.1000327.
Hatchwell, E. Is there a (host) genetic predisposition to progressive multifocal leukoencephalopathy? Frontiers in Immunology. May 2015; 6(216): pp. 1-5.
Hatchwell, et al. High rate of submicroscopic human genomic polymorphism detected by array CGH. Proceedings of XIX International Genetics Congress. Melbourne, Australia. Abstracts and Posters. 2003; 1.E.0092. pp. 168 and 319.
Hattersley, et al. What makes a good genetic association study? Lancet. Oct. 8, 2005;366(9493):1315-23.
Hay et al. Bacteriophage cloning and Escherichia coli expression of a human IgM Fab. Hum Antibodies Hybridomas 3(2):81-85 (1992).
He, et al. Analysis of de novo copy number variations in a family affected with autism spectrum disorders using high-resolution array-based comparative genomic hybridization. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. Jun. 2012;29(3):266-9. doi: 10.3760/cma.j.issn.1003-9406.2012.03.004. English abstract only.
Hegele, et al. “SNP Judgments and Freedom of Association”, Arterioscler. Thromb. Vase. Biol. 22 (2002): 1058-1061.
Helbig, et al. 15q13.3 microdeletions increase risk of idiopathic generalized epilepsy. Nat Genet. Feb. 2009;41(2):160-2. Epub Jan. 11, 2009.
Henchcliffe, et al., Mitochondrial biology and oxidative stress in Parkinson disease pathogenesis. Nat. Clin. Pract. Neurology, 2005;4(11):600-609.
Hicks et al., “Novel patterns of genome rearrangement and their association with survival in breast cancer,” Genome Res 16:1465-1479, 2006.
Hirschhorn, et al. A comprehensive review of genetic association studies. Genet Med. Mar.-Apr. 2002;4(2):45-61.
Hoffman, et al. Pharmacokinetic and pharmacodynamic aspects of gastroretentive dosage forms. Int J Pharm. Jun. 11, 2004;277(1-2):141-53.
Hoheisel, J. Microarray technology: beyond transcript profiling and genotype analysis. Nat Rev Genet. Mar. 2006;7(3):200-10.
Huang, et al. Whole genome DNA copy number changes identified by high density oligonucleotide arrays. Hum Genomics. May 2004;1(4):287-99.
Hudson, et al., Two-stage association study and meta-analysis of mitochondrial DNA variants in Parkinson disease. American Academy of Neurology. 2013;80: 2042-2048.
Human Genome CGH Microarrays—Details & Specifications, six printed pages from www.agilent.com, printed on May 20, 2015.
Hunt et al., Silent (Synonymous) SNPs: Should We Care About Them?, Methods in Molecular Biology. 2009; 578: 23-39.
Hunter, C. Genetics: a touch of elegance with RNAi. Curr Biol. Jun. 17, 1999;9(12):R440-2.
Huse, et al. Generation of a large combinatorial library of the immunoglobulin repertoire in phage lambda. Science. Dec. 8, 1989;246(4935):1275-81.
Hutvagner, et al. A microRNA in a multiple-turnover RNAi enzyme complex. Science. Sep. 20, 2002;297(5589):2056-60. Epub Aug. 1, 2002.
Iafrate, et al. Detection of large-scale variation in the human genome. Nat Genet. Sep. 2004;36(9):949-51. Epub Aug. 1, 2004.
International Preliminary Report on Patentability dated Mar. 16, 2017 for International Application No. PCT/US2015/000093.
International search report and written opinion dated Jan. 15, 2014 for PCT/US2013/062346.
International search report and written opinion dated Jan. 20, 2014 for PCT/US2013/059739.
International search report and written opinion dated Apr. 9, 2012 for PCT/US2011/001363.
International search report and written opinion dated Apr. 22, 2013 for PCT/US2012/063451.
International search report and written opinion dated Jul. 3, 2013 for PCT/IB2012/002498.
International search report and written opinion dated Dec. 18, 2019 for PCT/US2019/045721., (17 pages).
International Search Report dated Sep. 11, 2008 for PCT Application No. US2007/68183.
“Introducing Genome-Wide SNP Array 6.0 Pure performance & Genetic Power.” May 21, 2008. Available at http://www.genehk.com/news/doc/Genomics_genome-wide Human SNP Array 6.0.pdf. Accessed on Dec. 22, 2016.
Itsara, et al. Population analysis of large copy number variants and hotspots of human genetic disease. Am J Hum Genet. Feb. 2009;84(2):148-61. Epub Jan. 22, 2009.
Jain. Strategies and technologies for drug delivery systems. Trends in Pharmacological Sciences 19:155-157 (1998).
Jelcic, et al., Broadly neutralizing human monoclonal JC polyomavirus VP1-specific antibodies as candidate therapeutics for progressive multifocal leukoencephalopathy. Science Translational Medicine Sep. 23, 2015: vol. 7, Issue 306, pp. 306ra150 DOI: 10.1126/scitranslmed.aac8691.
Jonsson, et al., A mutation in APP protects against Alzheimer's disease and age-related cognitive decline. Nature. Aug. 2, 2012;488(7409):96-9. doi: 10.1038/nature11283.
Jorde, et al. Population genomics: a bridge from evolutionary history to genetic medicine. Hum. Mol. Genet. 2001; 10(20):2199-2207.
Juppner. Functional properties of the PTH/PTHrP receptor. Bone. Aug. 1995;17(2):Supplement 39S-42S.
Kalamatas, et al. Evaluation of serum JCV-Test and Index values in Natalizumab-treated individuals. American Neurological Association. 2015; S63:S507.
Kallioniemi, et al. Comparative genomic hybridization for molecular cytogenetic analysis of solid tumors. Science. Oct. 30, 1992;258(5083):818-21.
Kaminsky, et al., An evidence-based approach to establish the functional and clinical significance of copy number variants in intellectual and developmental disabilities. Genetics in medicine, 2011; 13(9): 777-784.
Karlin, et al. Applications and statistics for multiple high-scoring segments in molecular sequences. Proc Natl Acad Sci U S A. Jun. 15, 1993;90(12):5873-7.
Ketting, et al. Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans. Genes Dev. Oct. 15, 2001;15(20):2654-9.
Kevelam, et al., NUBPL mutations in patients with complex I deficiency and a distinct MRI pattern. Neurology. Apr. 23, 2013; 80(17): 1577-1583.
Kim, et al. Strategies for silencing human disease using RNA interference. Nat Rev Genet. Mar. 2007;8(3):173-84.
Kim, et al., Synthetic dsRNA dicer substrates enhance RNAi potency and efficacy. Nature biotechnology. 2005; 23(2): 222-226.
Kimchi-Sarfaty, et al. A “silent” polymorphism in the MDR1 gene changes substrate specificity. Science. Jan. 26, 2007;315(5811):525-8. Epub Dec. 21, 2006.
Klausner, et al. Novel gastroretentive dosage forms: evaluation of gastroretentivity and its effect on levodopa absorption in humans. Pharm Res. Sep. 2003;20(9):1466-73.
Klein, et al. Comparative genomic hybridization, loss of heterozygosity, and DNA sequence analysis of single cells. Proc Natl Acad Sci U S A. Apr. 13, 1999;96(8):4494-9.
Knight, et al. A cytogenetic abnormality and rare coding variants identify ABCA13 as a candidate gene in schizophrenia, bipolar disorder, and depression. Am J Hum Genet. Dec. 2009;85(6):833-46. doi: 10.1016/j.ajhg.2009.11.003.
Kohler, et al. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. Aug. 7, 1975;256(5517):495-7.
Kozbor, et al. The production of monoclonal antibodies from human lymphocytes. Immunol. Today. 1983; 4(3): 72-79.
Kraus, et al. Detection and isolation of novel protein-tyrosine kinase genes employing reduced stringency hybridization. Methods Enzymol. 1991;200:546-56.
Kumar, et al. A de novo 1p34.2 microdeletion identifies the synaptic vesicle gene RIMS3 as a novel candidate for autism. J Med Genet. Jun. 21, 2009. [Epub ahead of print].
Kumar, et al. Recurrent 16p11.2 microdeletions in autism. Hum Mol Genet. Feb. 15, 2008;17(4):628-38. Epub Dec. 21, 2007.
Kumar Kishore, et al., Genetics of parkinson disease and other movement disorders, Current opinion in neurology, Aug. 2012; 25(4):466-474.
Kurreck, J. Antisense technologies. Improvement through novel chemical modifications. Eur J Biochem. Apr. 2003;270(8):1628-44.
Kutyavin, et al. A novel endonuclease IV post-PCR genotyping system. Nucleic Acids Res. 2006;34(19):e128. Epub Sep. 29, 2006.
Kwoh et al. Transcription-based amplification system and detection of amplified human immunodeficiency virus type 1 with a bead-based sandwich hybridization format. PNAS USA 86(4):1173-1177 (1989).
Lakowicz, J. (1983) Principles of fluorescence spectroscopy. Plenum Press, New York.
Landegren, et al. A ligase-mediated gene detection technique. Science. Aug. 26, 1988;241(4869):1077-80.
Langston et al. Multisystem Lewy body disease and the other parkinsonian disorders. Nature Genetics 47(12):1378-1385 (2015).
Latchman, et al. Viral vectors for gene therapy in Parkinson's disease. Rev Neurosci. 2001;12(1):69-78.
Lavery, et al. Antisense and RNAi: powerful tools in drug target discovery and validation. Curr Opin Drug Discov Devel. Jul. 2003;6(4):561-9.
Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285-91 (2016).
Lerner. How to make a hybridoma. Yale J Biol Med. 54(5):387-402 (1981).
Liu et al: dbNSFP v3.0: A One-Stop Database of Functional Predictions and Annotations for Human Nonsynonymous and Splice-Site SNVs. Hum Mutat. 37(3):235-241. doi:10.1002/humu.22932 (2016).
Liu, Qing-Rong, et al. “Addiction molecular genetics: 639,401 SNP whole genome association identifies many “cell adhesion” genes.”American Journal of Medical Genetics Part B: Neuropsychiatric Genetics val. 141 (2006): pp. 918-925.
Lizardi, et al. Exponential amplification of recombinant-RNA hybridization probes. Nature Biotechnology 6.10 (1988): 1197-1202.
Lucentini, J. Gene Association Studies Typically Wrong. The Scientist, 18(24):20 (2004).
Maas, et al., Drug-associated progressive multifocal leukoencephalopathy: a clinical, radiological, and cerebrospinal fluid analysis of 326 cases. J Neurol. Oct. 2016;263(10):2004-21. doi: 10.1007/s00415-016-8217-x. Epub Jul. 11, 2016.
MacDonald, Jeffrey R et al. “The Database of Genomic Variants: a curated collection of structural variation in the human genome.” Nucleic acids research vol. 42,Database issue (2014): D986-92. doi:10.1093/nar/gkt958.
Maftei, et al. Interaction structure of the complex between neuroprotective factor humanin and Alzheimer's β-amyloid peptide revealed by affinity mass spectrometry and molecular modeling. J Pept Sci. Jun. 2012;18(6):373-82. doi: 10.1002/psc.2404. Epub Apr. 20, 2012.
Maniatis, et al. Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (1982).
Manolio, et al.Finding the missing heritability of complex diseases. Nature. Oct. 8, 2009;461(7265):747-53.
Margulies, et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature. Sep. 15, 2005;437(7057):376-80. Epub Jul. 31, 2005.
Marques, et al. A structural basis for discriminating between self and nonself double-stranded RNAs in mammalian cells. Nat Biotechnol. May 2006;24(5):559-65. Epub Apr. 30, 2006.
Marshall, et al. Structural variation of chromosomes in autism spectrum disorder. Am J Hum Genet. Feb. 2008;82(2):477-88. doi: 10.1016/j.ajhg.2007.12.009. Epub Jan. 17, 2008.
Martinez et al. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell 110(5):563-574 (2002).
Mast, et al. Invader assay for single-nucleotide polymorphism genotyping and gene copy number evaluation. Methods Mol Biol. 2006;335:173-86. Abstract only.
Matsuoka, et al. Humanin and the receptors for humanin. Mol Neurobiol. Feb. 2010;41(1):22-8. Epub Dec. 9, 2009.
May et al., Endometrial alterations in endometriosis: a systematic review of putative biomarkers. Hum. Reprod. Update, 17(5); 637-53:2011.
McCarroll, et al. Copy-number variation and association studies of human disease. Nat Genet. Jul. 2007;39(7 Suppl):S37-42.
McCarthy, et al. Microduplications of 16p11.2 are associated with schizophrenia. Nat Genet. Nov. 2009;41(11):1223-7. Epub Oct. 25, 2009.
McInnes, et al. A large-scale survey of the novel 15q24 microdeletion syndrome in autism spectrum disorders identifies an atypical deletion that narrows the critical region. Mol Autism. Mar. 19, 2010;1(1):5. doi: 10.1186/2040-2392-1-5.
McManus, et al. Gene silencing in mammals by small interfering RNAs. Nat Rev Genet. Oct. 2002;3(10):737-47.
Melis, et al., Drug-Induced Progressive Multifocal Leukoencephalopathy: A Comprehensive Analysis of the WHO Adverse Drug Reaction Database. CNS Drugs Oct. 2015, vol. 29, Issue 10, pp. 879-891.
Mills, Elizabeth A. et al., “Understanding Progressive Multifocal Leukoencephalapathy Risk in Multiple Sclerosis Patients Treated with Immunomodulatory Therapies: A Bird's Eye View”, Feb. 2018, Frontiers in Immunology, vol. 9, pp. 1-18.
Milner, et al. The cup runneth over: lessons from the ever-expanding pool of primary immunodeficiency diseases. Nature Reviews. Immunology, Sep. 2013; 13: pp. 635-648.
Mockler, et al. Applications of DNA tiling arrays for whole-genome analysis. Genomics. Jan. 2005;85(1):1-15.
Mohapatra, et al. Analyses of brain tumor cell lines confirm a simple model of relationships among fluorescence in situ hybridization, DNA index, and comparative genomic hybridization. Genes Chromosomes Cancer. Dec. 1997;20(4):311-9.
Mounsey et al., Mitochondrial Dysfunction in Parkinson's disease: Pathogenesis and neuroprotection. Parkinson's Disease, 2010: 18 pages.
Multiple Sclerosis. Special Interest Group Symposia 5. Session abstract. Sep. 27, 2015.
Mummidi et al., Evolution of human and non-human primate CC chemokine receptor 5 gene and mRNA. Journal of Biological Chemistry, 275(5):18946-18961 (2000).
Munoz-Amatriain et al., Distribution, functional impact, and origin mechanisms of copy number variation in the barley genome. Genome Biology, 2013; 14:r58 pp. 1-17.
Nakamura, et al., Codon usage tabulated from international DNA sequence databases: status for the year 2000. Nucleic Acids Res. 28(1):292 (2000).
Nakazawa et al. UV and skin cancer: specific p53 gene mutation in normal skin as a biologically relevant exposure measurement. PNAS USA 91(1):360-364 (1994).
Nalls, et al. Extended tracts of homozygosity identify novel candidate genes associated with late-onset Alzheimer's disease. Neurogenetics. Jul. 2009;10(3):183-90. doi: 10.1007/s10048-009-0182-4. Epub Mar. 7, 2009.
Nalls, et al. Imputation of sequence variants for identification of genetic risks for Parkinson's disease: a meta-analysis of genome-wide association studies. Lancet. Feb. 19, 2011;377(9766):641-9. doi: 10.1016/S0140-6736(10)62345-8. Epub Feb. 1, 2011.
National Center for Biotechnology Information. NCBI. Available at: https://www.ncbi.nlm.nih.gov/. Accessed on: Jun. 8, 2017.
Navabi et al.: Primary immunodeficiencies associated with eosinophilia. Allergy Asthma Clin Immunol. 12:27:1-12. doi:10.1186/s13223-016-0130-4. (2016).
NCBI. GenBank accession No. AL390798.3. Human chromosome 14 DNA sequence Bac R-21O19 of library RPCI-11 from chromosome 14 of Homo sapiens (Human), complete sequence. Apr. 28, 2011.
NCBI GenBank accession No. NG_12385.1. Mar. 27, 2012.
NCBI GenBank accession No. NM_207303.1. Apr. 20, 2004.
NCBI SNP Database rs201412882, ss491686165, Mar. 6, 2012 (National Library of Medicine, NIH, Bethesda, MD, USA).
NHLBI Exome Sequencing Project (ESP) Exome Variant Server. Available at: http://evs.gs.washington.edu/EVS/. Accessed on Jun. 8, 2017.
Nielsen et al. Sequence-selective recognition of DNA by strand displacement with a thymine-substituted polyamide. Science 254:1497-1500 (1991).
Nord, et al. Accurate and exact CNV identification from targeted high-throughput sequence data. BMC Genomics. Apr. 12, 2011;12:184.
Nouws et al., Assembly factors as a new class of disease genes for mitochondrial complex I deficiency: cause, pathology and treatment options. Brain, 2012;135:12-22.
Nykanen, et al. ATP requirements and small interfering RNA structure in the RNA interference pathway. Cell. Nov. 2, 2001;107(3):309-21.
O'Keefe, et al. High-resolution genomic arrays facilitate detection of novel cryptic chromosomal lesions in myelodysplastic syndromes. Exp Hematol. Feb. 2007;35(2):240-51.
Ozelius, et al. LRRK2 G2019S as a cause of Parkinson's disease in Ashkenazi Jews. N Engl J Med. Jan. 26, 2006;354(4):424-5.
Paciolla et al.: Rare mendelian primary immunodeficiency diseases associated with impaired NF-ΛB signaling. Genes Immun. 16(4):239-246. doi:10.1038/gene.2015.3 (2015).
Paisan-Ruiz Coro, et al., Parkinson's disease and low frequency alleles found together throughout LRRK2, Annals of human genetics. Jul. 2009. 73(4). 391-403.
Pang, et al. Towards a comprehensive structural variation map of an individual human genome. Genome Biol. 2010;11(5):R52. Epub May 19, 2010.
Pavlovic, et al., Progressive multifocal leukoencephalopathy: current treatment options and future perspectives. Ther Adv Neurol Disord 2015, vol. 8(6) 255-273 DOI: 10.1177/1756285615602832.
PCT/IB2018/000181 International Search Report and Written Opinion dated Nov. 6, 2018.
Peltz, et al. Targeting post-transcriptional control for drug discovery. RNA Biol. Jul.-Aug. 2009;6(3):329-34. Epub Jul. 7, 2009.
Pennisi. A closer look at SNPs suggests difficulties. Science. Sep. 18, 1998; 281(5384): 1787-1789.
Perkel, J. SNP genotyping: six technologies that keyed a revolution. Nature Methods. 2008;5:447-453.
Petrini, et al. The immunoglobulin heavy chain switch: structural features of gamma 1 recombinant switch regions. J Immunol. Mar. 15, 1987;138(6):1940-6.
Picard et al.: International Union of Immunological Societies: 2017 Primary Immunodeficiency Diseases Committee Report on Inborn Errors of Immunity. J Clin Immunol. 38(1):96-128 (2018).
Pinkel, et al. Comparative Genomic Hybridization. Annual Review of Genomics and Human Genetics, 6: 331-354 (2005).
Pinkel, et al. High resolution analysis of DNA copy number variation using comparative genomic hybridization to microarrays. Nat Genet. Oct. 1998;20(2):207-11.
Pinto, et al. Comprehensive assessment of array-based platforms and calling algorithms for detection of copy number variants. Nat Biotechnol. May 8, 2011;29(6):512-20. doi: 10.1038/nbt.1852.
Pinto, et al. Functional impact of global rare copy number variation in autism spectrum disorders. Nature. Jul. 15, 2010;466(7304):368-72. Epub Jun. 9, 2010.
Plasterk, et al. The silence of the genes. Curr Opin Genet Dev. Oct. 2000;10(5):562-7.
Poewe, et al., Parkinson disease. Nature Review:Disease Primers. Mar. 23, 2017.vol. 3, Article 17013: 1-21.
Pokharel, S. et al., High-Throughput Screening for Functional Adenosine to Inosine RNA Editing Systems. ACS Chem Biol. Dec. 15, 2006;1(12):761-5.
Pollack, et al. Microarray analysis reveals a major direct role of DNA copy number alteration in the transcriptional program of human breast tumors. Proc. Natl. Acad. Sci. 2002;99(20):12963-68.
Poplin et al.: A universal SNP and small-indel variant caller using deep neural networks. Nat Biotechnol. 36(10):983-987. doi:10.1038/nbt.4235 (2018).
Prasad, et al. A discovery resource of rare copy number variations in individuals with autism spectrum disorder. G3 (Bethesda). Dec. 2012;2(12):1665-85. doi: 10.1534/g3.112.004689. Epub Dec. 1, 2012.
Price, Alkes L et al. “Pooled association tests for rare variants in exon-resequencing studies.” American journal of human genetics vol. 86,6 (2010): 832-8. doi:10.1016/j.ajhg.2010.04.005.
Probe Reports from the NIH Molecular Libraries Program [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2010. Available from: https://www.ncbi.nlm.nih.gov/books/NBK47352/.
Provost, et al. Ribonuclease activity and RNA binding of recombinant human Dicer. EMBO J. Nov. 1, 2002;21(21):5864-74.
Purcell et al. “Postmortem brain abnormalities of the glutamate neurotransmitter system in autism” (Neurology, vol. 57 (2001) pp. 1618-1628).
Ragoussis, et al. Affymetrix GeneChip system: moving from research to the clinic. Expert Rev Mol Diagn. Mar. 2006;6(2):145-52.
Ramsey, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med. Nov. 3, 2011;365(18):1663-72.
Ray, et al., JC polyomavirus mutants escape antibody-mediated neutralization. Science Translational Medicine Sep. 23, 2015: vol. 7, Issue 306, pp. 306ra151 DOI: 10.1126/scitranslmed.aab1720.
Redon, et al. Global variation in copy number in the human genome. Nature. Nov. 23, 2006;444(7118):444-54.
Rees, et al. Isoform heterogeneity of the human gephyrin gene (GPHN), binding domains to the glycine receptor, and mutation analysis in hyperekplexia. J Biol Chem. Jul. 4, 2003;278(27):24688-96. Epub Apr. 8, 2003.
Reinke, T., MS Drug going generic without making waves. Medication Management. Jun. 2015.11 pages.
Remington “The Science and Practice of Pharmacy” (20th Ed., Lippincott Williams & Wilkins, Baltimore MD).
Revised American society for reproductive medicine classification of endometriosis: 1996. Fertility and Sterility. 67: 1997; 817-21.
Reynolds, et al. Rational siRNA design for RNA interference. Nat Biotechnol. Mar. 2004;22(3):326-30. Epub Feb. 1, 2004.
Risch, et al. A genomic screen of autism: evidence for a multilocus etiology. Am J Hum Genet. Aug. 1999;65(2):493-507.
Rodriguez-Revenga, et al. Structural variation in the human genome: the impact of copy number variants on clinical diagnosis. Genet Med. Sep. 2007;9(9):600-6.
Roohi, et al. Disruption of contactin 4 in three subjects with autism spectrum disorder. J Med Genet. Mar. 2009;46(3):176-82.
Saha, et al. Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell. Dec. 4, 2009;5(6):584-95.
Saiki, et al. Primer-directed enzymatic amplification of DNA with a thermostable DNA polymerase. Science. Jan. 29, 1988;239(4839):487-91.
Sambrook et al., Molecular Cloning: A Laboratory Manual (2 ed.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (1989).
Sanders, et al., Multiple Recurrent De Novo CNVs, including duplications of the 7q11.23 Williams Syndrome Region, Are Strongly Associated with Autism. Neuron. Jun. 9, 2011; 70:863-885.
Santa Cruz Human Genome Browser Gateway. 2017. Available at: http://genome.ucsc.edu/cgi-bin/hgGateway. Accessed on: Jun. 8, 2017.
Sauna, et al., Understanding the contribution of synonymous mutations to human disease.Nat Rev Genet. Aug. 31, 2011;12(10):683-91. doi: 10.1038/nrg3051.
Schapira. Causes of neuronal death in Parkinson's disease. Adv Neurol. 2001;86:155-62.
Schapira, et al. Mitochondrial complex I deficiency in Parkinson's disease. Lancet. Jun. 3, 1989;1(8649):1269.
Schapira. Mitochondrial complex I deficiency in Parkinson's disease. Adv Neurol. 1993;60:288-91.
Schroder, et al., Evaluation of RAG1 mutations in an adult with combined immunodeficiency and progressive multifocal leukoencephalopathy. Clinical Immunology 179 (2017) 1-7.
Schule, et al. Can cellular models revolutionize drug discovery in Parkinson's disease? Biochim Biophys Acta. Nov. 2009;1792(11):1043-51. Epub Sep. 3, 2009.
Schwab, et al., Therapy with natalizumab is associated with high JCV seroconversion and rising JCV index values. American Academy of Neurology. 2016. pp. 1-8.
Schwarz, et al. Asymmetry in the assembly of the RNAi enzyme complex. Cell. Oct. 17, 2003;115(2):199-208.
Sebat, et al. Large-Scale Copy Number Polymorphism in the Human Genome. Science, 305:525-528 (2004).
Sebat, et al. Strong association of de novo copy number mutations with autism. Science. Apr. 20, 2007;316(5823):445-9.
Serana, et al., Immunological biomarkers identifying natalizumab-treated multiple sclerosis patients at risk of progressive multifocal leukoencephalopathy. Journal of Neuroimmunology, 277 (2014) 6-12.
Seshan Ve and Olshen A (2017). DNAcopy: DNA copy number data analysis. R package version 1.50.1. Available at: http://www.bioconductor.org/packages/release/bioc/html/DNAcopy.html.
Sharp. RNA interference—2001. Genes Dev 15(5):485-490 (2001).
Sheftel, et al., Human Ind1, an Iron-Sulfur Cluster Assembly Factor for Respiratory Complex I. Molecular and Cellular Biology, Nov. 2009, p. 6059-6073.
Shi, Y. Mammalian RNAi for the masses. Trends Genet. Jan. 2003;19(1):9-12.
Shuey, et al. RNAi: gene-silencing in therapeutic intervention. Drug Discov Today. Oct. 15, 2002;7(20):1040-6.
Sidransky, et al., Multicenter analysis of glucocerebrosidase mutations in Parkinson's disease.N Engl J Med. Oct. 22, 2009;361(17):1651-61. doi: 10.1056/NEJMoa0901281.
Sidrasky, E. Gaucher Disease: Insights from a Rare Mendelian Disorder. Discov Med. Author manuscript; available in PMC Aug. 22, 2014.
Simon-Sanchez, et al. Genome-wide association study reveals genetic risk underlying Parkinson's disease. Nat Genet. Dec. 2009;41(12):1308-12. doi: 10.1038/ng.487. Epub Nov. 15, 2009. with supplemental information.
Siolas, et al. Synthetic shRNAs as potent RNAi triggers. Nat Biotechnol. Feb. 2005;23(2):227-31. Epub Dec. 26, 2004.
Smith, et al. A high-density admixture map for disease gene discovery in african americans. Am J Hum Genet. May 2004;74(5):1001-13. Epub Apr. 14, 2004.
Snijders, et al. Assembly of microarrays for genome-wide measurement of DNA copy number. Nat Genet. Nov. 2001;29(3):263-4.
Snijders, et al. BAC microarray-based comparative genomic hybridization. Methods Mol Biol. 2004;256:39-56.
Snijders, et al. Mapping segmental and sequence variations among laboratory mice using BAC array CGH. Genome Res. Feb. 2005;15(2):302-11.
Soni, et al. Progress toward ultrafast DNA sequencing using solid-state nanopores. Clin Chem. Nov. 2007;53(11):1996-2001. Epub Sep. 21, 2007.
Spelman, et al., Comparative efficacy of first-line natalizumab vs IFN-B or glatiramer acetate in relapsing MS. Neurology clinical practice. 2016. pp. 102-115.
Stark, et al. De novo 325 kb microdeletion in chromosome band 10q25.3 including ATRNL1 in a boy with cognitive impairment, autism and dysmorphic features. Eur J Med Genet. Sep.-Oct. 2010;53(5):337-9. doi: 10.1016/j.ejmg.2010.07.009. Epub Jul. 27, 2010.
Stefansson, et al. Large recurrent microdeletions associated with schizophrenia. Nature. Sep. 11, 2008;455(7210):232-6.
Stephens, et al. Antisense oligonucleotide therapy in cancer. Curr Opin Mol Ther. Apr. 2003;5(2):118-22.
Streubel, et al. Gastroretentive drug delivery systems. Expert Opin Drug Deliv. Mar. 2006;3(2):217-33.
String. Search single protein by name/identifier. String consortium 2017. Available at:https://string-db.org/.
Sudhof. Neuroligins and neurexins link synaptic function to cognitive disease. Nature. Oct. 16, 2008;455(7215):903-11. doi: 10.1038/nature07456.
Summary of NRSP-8 Accomplishments: 2003-2008. Available at http://www.lgu.umd.edu/lgu_v2/pages/attachs/9956_Attach1%20%202003-08%20ACCOMPLISHMENTS.doc. Published on Feb. 9, 2008. (6 pages).
Sundqvist, et al., JC Polyomavirus infection is strongly controlled by human leucocyte antigen class II variants. PLOS Pathog, 2014;10(4): e1004084. doi:10.1371/journal.ppat.1004084.
Suryawanshi, S., et al., “Plasma microRNAs as novel biomarkers for endometriosis and endometriosis-associated ovarian cancer”, Clin Cancer Res., 19(5), (Mar. 1, 2013), 1213-24.
Szoka et al. Procedure for preparation of liposomes with large internal aqueous space and high capture by reverse-phase evaporation. PNAS. 1978;75:4194-4198.
Tabara, et al. The dsRNA binding protein RDE-4 interacts with RDE-1, DCR-1, and a DExH-box helicase to direct RNAi in C. elegans. Cell. Jun. 28, 2002;109(7):861-71.
Tabuchi, et al. A neuroligin-3 mutation implicated in autism increases inhibitory synaptic transmission in mice. Science. Oct. 5, 2007;318(5847):71-6. Epub Sep. 6, 2007.
Tam, et al. The role of DNA copy number variation in schizophrenia. Biol Psychiatry. Dec. 1, 2009;66(11):1005-12. doi: 10.1016/j.biopsych.2009.07.027. Epub Sep. 12, 2009.
Tenisch, et al., Massive and exclusive pontocerebellar damage in mitochondrial disease and NUBPL mutations.Neurology. Jul. 24, 2012;79(4):391. doi: 10.1212/WNL.0b013e3182611232.
Teo, et al. Statistical challenges associated with detecting copy number variations with next-generation sequencing. Bioinformatics. Aug. 31, 2012.
The 1000 Genomes project consortium. An integrated map of genetic variation from 1,092 human genomes. 56 | Nature | vol. 491 | Nov. 1, 2012.
The International Schizophrenia Consortium. Rare chromosomal deletions and duplications increase risk of schizophrenia. Nature. Sep. 11, 2008;455(7210):237-41. Epub Jul. 30, 2008.
The Merck Manual of Diagnosis and Therapy, 18th Edition, published by Merck Research Laboratories, 2006 (ISBN 0-911910-18-2).
Thijssen et al.: Mutations in CDCA7 and HELLS cause immunodeficiency-centromeric instability-facial anomalies syndrome. Nat Commun. 6:7870:1-8. doi:10.1038/ncomms8870 (2015).
Thompson. Applications of antisense and siRNAs during preclinical drug development. Drug Discov Today. Sep. 1, 2002;7(17):912-7.
Thorpe, et al. Improved antitumor effects of immunotoxins prepared with deglycosylated ricin A-chain and hindered disulfide linkages. Cancer Res. Nov. 15, 1988;48(22):6396-403.
Tonk et al.: Assessment of pharmacogenetic tests: presenting measures of clinical validity and potential population impact in association studies. Pharmacogenomics J. 17(4):386-392. doi:10.1038/tpj.2016.34 (2017).
Tsujita et al.: Phosphatase and tensin homolog (PTEN) mutation can cause activated phosphatidylinositol 3-kinase δ[Delta] syndrome-like immunodeficiency. J Allergy Clin Immunol. 138(6):1672-1680.e10. doi:10.1016/j.jaci.2016.03.055 (2016).
Tucker et al. Next-generation sequencing in molecular diagnosis: NUBPL mutations highlight the challenges of variant detection and interpretation. Human mutation, 2012; 33(2):411-418.
UK Parkinson's Disease Consortium et al., Dissection of the genetics of parkinson's disease identifies an additional association 5′ of SNCA and multiple associated haplotypes at 17q21. Human Molecular genetics. Jan. 15, 2011; 20(2): 345-353.
Urnov, et al. Genome editing with engineered zinc finger nucleases. Nat Rev Genet. Sep. 2010;11(9):636-46.
Van Der Kolk, et al., Progressive multifocal leukoencephalopathy in an immunocompetent patient. Annals of Clinical and Translational Neurology 2016; 3(3): 226-232.
Van Goor, et al. Correction of the F508del-CFTR protein processing defect in vitro by the investigational drug VX-809. Proc Natl Acad Sci U S A. Nov. 15, 2011;108(46):18843-8. Epub Oct. 5, 2011.
Van Goor, et al. Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Proc Natl Acad Sci U S A. Nov. 3, 2009;106(44):18825-30. Epub Oct. 21, 2009.
Vaughan, et al. Genetics of Parkinsonism: a review. Ann Hum Genet. Mar. 2001;65(Pt 2):111-26.
Veensra-Vanderweele, et al. Networking in autism: leveraging genetic, biomarker and model system findings in the search for new treatments. Neuropsychopharmacology. Jan. 2012;37(1):196-212. doi: 10.1038/npp.2011.185. Epub Sep. 21, 2011.
Vickers, et al. Efficient reduction of target RNAs by small interfering RNA and RNase H-dependent antisense agents. A comparative analysis. J Biol Chem. Feb. 28, 2003;278(9):7108-18. Epub Dec. 23, 2002.
Vissers, et al. Array-based comparative genomic hybridization for the genomewide detection of submicroscopic chromosomal abnormalities. Am. J. Hum. Genet. 2003; 73:1261-70.
Vissers, et al. Identification of disease genes by whole genome CGH arrays. Hum Mol Genet. Oct. 15, 2005;14 Spec No. 2:R215-223.
Walker, et al. Genetic analysis of attractin homologs. Genesis. 2007; 45(12):744-756.
Walsh, et al. Detection of inherited mutations for breast and ovarian cancer using genomic capture and massively parallel sequencing. Proc Natl Acad Sci U S A. Jul. 13, 2010;107(28):12629-33. doi: 10.1073/pnas.1007983107. Epub Jun. 28, 2010.
Walsh, et al. Spectrum of mutations in BRCA1, BRCA2, CHEK2, and TP53 in families at high risk of breast cancer. JAMA. Mar. 22, 2006;295(12):1379-88.
Walters, et al. A novel highly penetrant form of obesity due to deletions on chromosome 16p11.2. Nature. Feb. 4, 2010;463(7281):671-5.
Wang, et al. Antisense anticancer oligonucleotide therapeutics. Curr Cancer Drug Targets. Nov. 2001;1(3):177-96.
Weiss, et al. Association between microdeletion and microduplication at 16p11.2 and autism. N Engl J Med. Feb. 14, 2008;358(7):667-75.
Westmark, C. What's hAPPening at synapses? The role of amyloid β-protein precursor and β-amyloid in neurological disorders. Mol Psychiatry. Aug. 28, 2012. doi: 10.1038/mp.2012.122.
Wilson, et al. DNA copy-number analysis in bipolar disorder and schizophrenia reveals aberrations in genes involved in glutamate signaling. Hum Mol Genet. Mar. 1, 2006;15(5):743-9. Epub Jan. 24, 2006.
Xia, et al. siRNA-mediated gene silencing in vitro and in vivo. Nat Biotechnol. Oct. 2002;20(10):1006-10. Epub Sep. 16, 2002.
Xiao, et al., Identification and optimization of small-molecule agonists of the human relaxin hormone receptor RXFP1. Nat Commun. 2013;4:1953. doi:10.1038/ncomms2953.
Xie, et al. CNV-seq, a new method to detect copy number variation using high-throughput sequencing. BMC Bioinformatics. Mar. 6, 2009;10:80.
Yusa, et al. Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells. Nature. Oct. 12, 2011;478(7369):391-4. doi: 10.1038/nature10424.
Zapala, et al. Humanins, the neuroprotective and cytoprotective peptides with antiapoptotic and anti-inflammatory properties. Pharmacol Rep. Sep.-Oct. 2010;62(5):767-77.
Zeng, Li, et al. “A novel splice variant of the cell adhesion molecule contactin 4 (CNTN4) is mainly expressed in human brain.” Journal of human genetics val. 47 (2002): pp. 497-499.
Zerbe, et al., Progressive Multifocal Leukoencephalopathy in Primary Immune Deficiencies: Stat1 Gain of Function and Review of the Literature. IDSA 2016. pp. 1-9.
Zhang, et al. Copy number variation in human health, disease, and evolution. Annu Rev Genomics Hum Genet. 2009;10:451-81. doi: 10.1146/annurev.genom.9.081307.164217.
Zhang, et al. Detection of copy number variation from array intensity and sequencing read depth using a stepwise Bayesian model. BMC Bioinformatics. Oct. 31, 2010;11:539.
Zhao et al. (eds), Bacterial Artificial Chromosomes: Methods Protocols Methods in Molecular Biology, Humana Press, 2004.
Zhou et al.: Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet. 48(1):67-73. doi:10.1038/ng.3459 (2016).
Ziats, et al. Expression profiling of autism candidate genes during human brain development implicates central immune signaling pathways. PLoS One. 2011;6(9):e24691. doi:10.1371/journal.pone.0024691. Epub Sep. 15, 2011.
Chen et al.: Genetic polymorphism of 3′ untranslated region of zeta-chain associated protein kinase 70 kDa in southern Taiwanese patients with rheumatoid arthritis. Clin Rheumatol. 35(3):747-750. doi:10.1007/s10067-015-3044-5 (2016).
Ghadery et al.: Microglial activation in Parkinson's disease using [18F]-FEPPA. J Neuroinflammation. 14(1):8:1-9. doi:10.1186/s12974-016-0778-1 (2017).
Pawlik et al.: IL17A and IL17F gene polymorphisms in patients with rheumatoid arthritis. BMC Musculoskelet Disord. 17:208:1-6. doi:10.1186/s12891-016-1064-1 (2016).
Related Publications (1)
Number Date Country
20230265519 A1 Aug 2023 US
Provisional Applications (2)
Number Date Country
62524324 Jun 2017 US
62454676 Feb 2017 US
Divisions (2)
Number Date Country
Parent 17141885 Jan 2021 US
Child 18185153 US
Parent 16245849 Jan 2019 US
Child 17141885 US
Continuations (1)
Number Date Country
Parent 15639591 Jun 2017 US
Child 16245849 US