Mammalian Genes Involved in Infection

Abstract
The present invention relates to nucleic acid sequences and cellular proteins encoded by these sequences that are involved in infection or are otherwise associated with the life cycle of one or more pathogens.
Description
FIELD OF THE INVENTION

The present invention relates to nucleic acid sequences and cellular proteins encoded by these sequences that are involved in infection or are otherwise associated with the life cycle of one or more pathogens, such as a virus, a bacteria, a fungus or a parasite. The invention also relates to modulators of nucleic acid sequences and cellular proteins encoded by these sequences that are involved in infection or are otherwise associated with the life cycle of a pathogen.


BACKGROUND

Infectious diseases affect the health of people and animals around the world, causing serious illness and death. Black Plague devastated the human population in Europe during the middle ages. Pandemic flu killed millions of people in the 20th century and is a threat to reemerge.


Some of the most feared, widespread, and devastating human diseases are caused by viruses that interfere with normal cellular processes. These include influenza, poliomyelitis, smallpox, Ebola, yellow fever, measles and AIDS, to name a few. Viruses are also responsible for many cases of human disease including encephalitis, meningitis, pneumonia, hepatitis and cervical cancer, warts and the common cold. Furthermore, viruses causing respiratory infections, and diarrhea in young children lead to millions of deaths each year in less-developed countries. Also, a number of newly emerging human diseases such as SARS are caused by viruses. In addition, the threat of a bioterrorist designed pathogen is ever present.


While vaccines have been effective to prevent certain viral infections, relatively few vaccines are available or wholly effective, have inherent risks and tend to be specific for particular conditions. Vaccines are of limited value against rapidly mutating viruses and cannot anticipate emerging viruses or new bioterrorist designed viruses. Currently there is no good answer to these threats.


Traditional treatments for viral infection include pharmaceuticals aimed at specific virus derived proteins, such as HIV protease or reverse transcriptase, or the administration of recombinant (cloned) immune modulators (host derived), such as the interferons. However, the vast majority of viruses lack an effective drug. Those drugs that exist have several limitations and drawbacks that including limited effectiveness, toxicity, and high rates of viral mutations which render antiviral pharmaceuticals ineffective. Thus, an urgent need exists for alternative treatments for viruses and other infectious diseases, and methods of identifying new drugs to combat these threats.


SUMMARY OF THE INVENTION

The present invention provides genes and gene products set forth in Table 1 that are involved in infection by one or more pathogens such as a virus, a parasite, a bacteria or a fungus, or are otherwise associated with the life cycle of a pathogen. Also provided are methods of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more of these genes or gene products set forth in Table 1. Also provided are methods of decreasing infection by a pathogen in a subject by administering an agent that decreases the expression and/or activity of the genes or gene products set forth in Table 1. Further provided are methods of identifying an agent that decreases infection by a pathogen.







DETAILED DESCRIPTION OF THE INVENTION

The present invention may be understood more readily by reference to the following detailed description of preferred embodiments of the invention and the Examples included therein.


Before the present compounds, compositions and/or methods are disclosed and described, it is to be understood that this invention is not limited to specific nucleic acids, specific polypeptides, or to particular methods, as such may, of course, 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 be limiting.


As used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. The term “or” refers to a single element of stated alternative elements or a combination of two or more elements, unless the context clearly indicates otherwise. As used herein, “comprises” means “includes.” Thus, “comprising A or B,” means “including A, B, or A and B,” without excluding additional elements.


Ranges may be expressed herein as from “about” one particular value, and/or to “about” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint.


“Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, the phrase “optionally obtained prior to treatment” means obtained before treatment, after treatment, or not at all.


As used throughout, by “subject” is meant an individual. Preferably, the subject is a mammal such as a primate, and, more preferably, a human. Non-human primates include marmosets, monkeys, chimpanzees, gorillas, orangutans, and gibbons, to name a few. The term “subject” includes domesticated animals, such as cats, dogs, etc., livestock (for example, cattle, horses, pigs, sheep, goats, etc.), laboratory animals (for example, ferret, chinchilla, mouse, rabbit, rat, gerbil, guinea pig, etc.) and avian species (for example, chickens, turkeys, ducks, pheasants, pigeons, doves, parrots, cockatoos, geese, etc.). The subjects of the present invention can also include, but are not limited to fish (for example, zebrafish, goldfish, tilapia, salmon and trout), amphibians and reptiles.


In the present application, the genes listed in Table 1 are host genes involved in viral infection. All of the host genes involved in viral infection, set forth in Table 1, were identified using gene trap methods that were designed to identify host genes that are necessary for viral infection or growth, but nonessential for cellular survival. These gene trap methods are set forth in the Examples as well as in U.S. Pat. No. 6,448,000 and U.S. Pat. No. 6,777,177. U.S. Pat. Nos. 6,448,000 and 6,777,177 and are both incorporated herein in their entireties by this reference.


As used herein, a gene “nonessential for cellular survival” means a gene for which disruption of one or both alleles results in a cell viable for at least a period of time which allows viral replication to be decreased or inhibited in a cell. Such a decrease can be utilized for preventative or therapeutic uses or used in research. A gene necessary for pathogenic infection or growth means the gene product of this gene, either protein or RNA, secreted or not, is necessary, either directly or indirectly in some way for the pathogen to grow. As utilized throughout, “gene product” is the RNA or protein resulting from the expression of a gene listed in Table 1.


The nucleic acids of these genes and their encoded proteins can be involved in all phases of the viral life cycle including, but not limited to, viral attachment to cellular receptors, viral infection, viral entry, internalization, disassembly of the virus, viral replication, genomic integration of viral sequences, transcription of viral RNA, translation of viral mRNA, transcription of cellular proteins, translation of cellular proteins, trafficking, proteolytic cleavage of viral proteins or cellular proteins, assembly of viral particles, budding, cell lysis and egress of virus from the cells.


Although the genes set forth herein were identified as cellular genes involved in viral infection, as discussed throughout, the present invention is not limited to viral infection. Therefore, any of these nucleic acid sequences and the proteins encoded by these sequences can be involved in infection by any infectious pathogen such as a bacteria, a fungus or a parasite which includes involvement in any phase of the infectious pathogen's life cycle.


As utilized herein, when referring to any of the genes in Table 1 for example, and not to be limiting, AHR, this includes any AHR gene, AHR gene product, for example, an AHR nucleic acid (DNA or RNA) or AHR protein, from any organism that retains at least one activity of AHR and can function as an AHR nucleic acid or protein utilized by a pathogen. For example, the nucleic acid or protein sequence can be from or in a cell in a human, a non-human primate, a mouse, a rat, a cat, a dog, a chimpanzee, a horse, a cow, a pig, a sheep, a guinea pig, a rabbit, a zebrafish, a chicken, to name a few.


As used herein, a gene is a nucleic acid sequence that encodes a polypeptide under the control of a regulatory sequence, such as a promoter or operator. The coding sequence of the gene is the portion transcribed and translated into a polypeptide (in vivo, in vitro or in situ) when placed under the control of an appropriate regulatory sequence. The boundaries of the coding sequence can be determined by a start codon at the 5′ (amino) terminus and a stop codon at the 3′ (carboxyl) terminus. If the coding sequence is intended to be expressed in a eukaryotic cell, a polyadenylation signal and transcription termination sequence can be included 3′ to the coding sequence.


Transcriptional and translational control sequences include, but are not limited to, DNA regulatory sequences such as promoters, enhancers, and terminators that provide for the expression of the coding sequence, such as expression in a host cell. A polyadenylation signal is an exemplary eukaryotic control sequence. A promoter is a regulatory region capable of binding RNA polymerase and initiating transcription of a downstream (3′ direction) coding sequence. Additionally, a gene can include a signal sequence at the beginning of the coding sequence of a protein to be secreted or expressed on the surface of a cell. This sequence can encode a signal peptide, N-terminal to the mature polypeptide, which directs the host cell to translocate the polypeptide.


Table 1 (column 2) provides one or more aliases for each of the genes set forth herein. Therefore, it is clear that when referring to a gene, this also includes known alias(es) and any aliases attributed to the genes listed in Table 1 in the future. Table 1 also provides the Entrez Gene numbers for the human genes set forth herein. The information provided under the Entrez Gene numbers listed in Table 1 is hereby incorporated entirely by this reference. One of skill in the art can readily obtain this information from the National Center for Biotechnology Information at the National Library of Medicine (http://www.ncbi.nlm.nih.gov/entrez/query.fegi?db=gene). By accessing Entrez Gene, one of skill in the art can readily obtain information about every gene listed in Table 1, such as the genomic location of the gene, a summary of the properties of the protein encoded by the gene, expression patterns, function, information on homologs of the gene as well as numerous reference sequences, such as the genomic, mRNA and protein sequences for each gene. Therefore, one of skill in the art can readily obtain sequences, such as genomic, mRNA and protein sequences by accessing information available under the Entrez Gene number provided for each gene. Thus, all of the information readily obtained from the Entrez Gene Nos. set forth herein is also hereby incorporated by reference in its entirety.


Also provided in Table 1 are the GenBank Accession Nos. for the human mRNA sequences and the GenBank Accession Nos. for the human protein sequences if available. For certain non-protein coding genes, a non-coding RNA is provided, for example, for SNORA molecules. The nucleic acid sequences and protein sequences provided under the GenBank Accession Nos. mentioned herein are hereby incorporated in their entireties by this reference. One of skill in the art would know that the nucleotide sequences provided under the GenBank Accession Nos. set forth herein can be readily obtained from the National Center for Biotechnology Information at the National Library of Medicine (http://www.hcbi.nlm.nih.gov/entrez/query.fcgi?db=nucleotide). It is understood that in any coding sequence, a T can be replaced by a U to obtain an RNA sequence for each gene.


Similarly, the protein sequences set forth herein can be readily obtained from the National Center for Biotechnology Information at the National Library of Medicine (http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=protein). The nucleic acid sequences and protein sequences provided under the GenBank Accession Nos. mentioned herein are hereby incorporated in their entireties by this reference.


These examples are not meant to be limiting as one of skill in the art would know how to obtain additional sequences for the genes and gene products listed in Table 1 from other species by accessing GenBank (Benson et al. Nucleic Acids Res. 2004 January 1; 32 (Database Issue); D23-D26), the EMBL Database (Stoesser et al. (2000) Nucleic Acids Res, 28, 19-23) or other sequence databases. One of skill in the art would also know how to align the sequences disclosed herein with sequences from other species in order to determine similarities and differences between the sequences set forth in Table 1 and related sequences, for example, by utilizing BLAST. As set forth herein, a nucleic acid sequence for any of the genes set forth in Table 1 can be a full-length wild-type (or native) sequence, a genomic sequence, a variant (for example, an allelic variant or a splice variant), a nucleic acid fragment, a homolog or a fusion sequence that retains the activity of the gene utilized by the pathogen or its encoded gene product.
























Human GenBank








Human
Accession
Human GenBank





Function/
Chrom.
No. for coding sequence/
Accession No.


Gene
Alias
Definition
Association
Location
mRNA (SEQ ID NO:)
for protein (SEQ ID NO:)
Entrez Gene No.






















MTAP
MSAP; c86fus
methylthio-adenosine
Encodes an enzyme
9p21
NM_002451.3 (1)
NP_002442.2 (2)
4507




phosphorylase
that plays a major





role in polyamine





metabolism and is





important for the





salvage of both





adenine and





methionine.


AHR
bHLHe76
aromatic hydrocarbon
Encodes a ligand-
7p15
NM_001621.4 (3)
NP_001612.1 (4)
196




receptor
activated





transcription factor





involved in the





regulation of





biological responses





to planar aromatic





hydrocarbons.


AK5
AK6, MGC33326, ATP-
adenylate kinase 6
Encodes a member of
1p31
NM_012093.2 (5)
NP_036225.2 (6)
26289



AMP transphosphorylase;

the adenylate kinase

NM_174858.1 (7)
NP_777283.1 (8)



OTTHUMP00000011354;

family, which is



OTTHUMP00000011355

involved in regulating





the adenine





nucleotide





composition within a





cell.


AMOTL2
LCCP
angiomotin-like protein 2
Related to
3q21-q22
NM_016201.2 (9)
NP_057285.3
51421





angiomotin and is a


(10)





member of the motins





protein family.


ANKMY2
ZMYND20;
ankyrin repeat and MYND
Uknown
7p21
NM_020319.2
NP_064715.1
57037



DKFZp564O043
domain containing 2


(11)
(12)


ANXA4
ANX4; PIG28; ZAP36;
annexin A4
Belongs to the
2p13
NM_001153.3
NP_001144.1
307



MGC75105;

annexin family of

(13)
(14)



DKFZp686H02120

calcium-dependent





phospholipid binding





proteins.


ARL6IP5
JWA; jmx; hp22; PRAF3;
ADP-ribosylation-like factor
Encoded protein of
3p14
NM_006407.3
NP_006398.1
10550



DERP11; HSPC127;
6 interacting protein 5
this gene may be

(15)
(16)



addicsin; GTRAP3-18

associated with the





cytoskeleton.


ARSA
MLD
arylsulfatase A
Hydrolyzes
22q13.31-qter;
NM_000487.4
NP_000478.2
410





cerebroside sulfate to
22q13.33
(17)
(18)





cerebroside and

NM_001085425.1
NP_001078894.1





sulfate.

(19)
(20)







NM_001085426.1
NP_001078895.1







(21)
(22)







NM_001085427.1
NP_001078896.1







(23)
(24)







NM_001085428.1
NP_001078897.1







(25)
(26)


ATOH8
HATH6; bHLHa21;
atonal homolog 8
Unknown
2p11.2
NM_032827.6
NP_116216.2
84913



FLJ14708; FLJ38730



(27)
(28)


ATP6V1A
HO68; VA68; VPP2;
ATPase, H+ transporting,
Encodes a component
3q13.2-q13.31
NM_001690.2
NP_001681.2
523



Vma1; ATP6A1;
lysosomal 70 kDa, V1 subunit A
of vacuolar ATPase

(29)
(30)



ATP6V1A1

(V-ATPase).


BPNT1
PIP
3′(2′), 5′-bisphosphate
A member of a
1q41
NM_006085.4
NP_006076.4
10380




nucleotidase 1
magnesium-

(31)
(32)





dependent





phosphomonoesterase





family.


C11ORF54
PTD012
chromosome 11 open reading
Unknown
11q21
NM_014039.2
NP_054758.2
28970




frame 54


(33)
(34)


C17ORF75
NJMU-R1
chromosome 17 open reading
Unknown
17q11.2
NM_022344.2
NP_071739.2
64149




frame 75


(35)
(36)


C18ORF32
FLJ23458
chromosome 18 open reading
Unknown
18q21.1
NM_001035005.2
NP_001030177.1
497661




frame 32


(37)
(38)


C1ORF116
SARG; MGC2742;
chromosome 1 open reading
Unknown
1q32.1-q32.2
NM_001083924.1
NP_001077393.1
79098



MGC4309; FLJ36507;
frame 116


(39)
(40)



DKFZp666H2010



NM_023938.5
NP_076427.2







(41)
(42)


C6ORF176
bA142J11.1
chromosome 6 open reading
Unknown
6q27
NR_026861.1

90632




frame 176


(43)







NR_026860.1







(44)


C6ORF62
XTP12; FLJ12619;
chromosome 6 open reading
Unknown
6p22.3
NM_030939.4
NP 112201.1
81688



dJ30M3.2; Nbla00237;
frame 62


(45)
(46)



DKFZp564G182


CBLN2

cerebellin 2 precursor
Unknown
18q22.3
NM_182511.3
NP_872317.1
147381







(47)
(48)


CCDC57
FLJ00130; FLJ23754;
coiled-coil domain containing
Unknown
17q25.3
NM_198082.2
NP_932348.2
284001



FLJ43953; MGC102869
57


(49)
(50)


CDH9
MGC125386
cadherin 9, type 2 (T1-
A type II classical
5p14
NM_016279.3
NP_057363.3
1007




cadherin)
cadherin from the

(51)
(52)





cadherin superfamily,





mediates calcium-





dependent cell-cell





adhesion.


CEP152
KIAA0912
centrosomal protein 152 kDa
Unknown
15q21.1
NM_014985.2
NP_055800.2
22995







(53)
(54)


CKAP2L
FLJ40629; MGC39683
cytoskeleton associated
Unknown
2q13
NM_152515.3
NP_689728.3
150468




protein 2-like


(55)
(56)


CLTC
Hc; CHC; CHC17; CLH-
clathrin, heavy chain
A major protein
17q11-qter
NM_004859.3
NP_004850.1
1213



17; CLTCL2; KIAA0034

component of the

(57)
(58)





cytoplasmic face of





intracellular





organelles, called





coated vesicles and





coated pits.


CLUL1
RA337M
clusterin-like 1 (retinal)
Unknown
18p11.32
NM_014410.4
NP_055225.1
27098







(59)
(60)







NM_199167.1
NP_954636.1







(61)
(62)


CNTN5
NB-2; HNB-2s;
contactin 5
A member of the
11q21-q22.2
NM_014361.2
NP_055176.1
53942



MGC163491

immunoglobulin

(63)
(64)





superfamily, may

NM_175566.1
NP_780775.1





play a role in the

(65)
(66)





formation of axon





connections in the





developing nervous





system.


CRYZ
FLJ41475;
crystallin, zeta (quinone
A taxon-specific
1p31-p22
NM_001130042.1
NP_001123514.1
1429



DKFZp779E0834
reductase)
crystallin protein

(67)
(68)





which has NADPH-

NM_001130043.1
NP_001123515.1





dependent quinone

(69)
(70)





reductase activity

NM_001134759.1
NP_001128231.1





distinct from other

(71)
(72)





known quinone

NM_001889.3
NP_001880.2





reductases.

(73)
(74)


DAB2
DOC2; DOC-2;
disabled homolog 2, mitogen-
A putative mitogen-
5p13
NM_001343.2
NP_001334.2
1601



FLJ26626
responsive phosphoprotein
responsive

(75)
(76)




(Drosophila)
phosphoprotein.


DARS2
LBSL; ASPRS;
aspartyl-tRNA synthetase 2,
Protein
1q25.1
NM_018122.4
NP_060592.2
55157



FLJ10514; MT-ASPRS;
mitochondrial
aminoacylates

(77)
(78)



RP3-383J4.2

aspartyl-tRNA.


DDIT4
Dig2; REDD1, REDD-1,
DNA-damage-inducible
Unknown
10pter-q26.12
NM_019058.2
NP_061931.1
54541



RTP801; FLJ20500;
transcript 4


(79)
(80)



RP11-442H21.1


DDX42
RHELP; RNAHP;
DEAD (Asp-Glu-Ala-Asp)
Implicated in a
17q23.3
NM_007372.2
NP_031398.2
11325



SF3b125; FLJ43179
box polypeptide 42
number of cellular

(81)
(82)





processes involving

NM_203499.1
NP_987095.1





alteration of RNA

(83)
(84)





secondary structure





such as translation





initiation, nuclear and





mitochondrial





splicing, and





ribosome and





spliceosome





assembly.


DNAH2
DNHD3; DNAHC2,
dynein, axonemal, heavy
Microtubule-
17p13.1
NM_020877.2
NP_065928.2
146754



FLJ46675; KIAA1503
chain 2
associated motor

(85)
(86)





protein complex.


DUSP5
DUSP; HVH3
dual specificity phosphatase 5
Negatively regulates
10q25
NM_004419.3
NP_004410.3
1847





members of the

(87)
(88)





mitogen-activated





protein (MAP) kinase





superfamily,





specifically ERK1.


EBNA1BP2
P40, EBP2, NOBP
EBNA1 binding protein 2
Unknown
1p35-p33
NM_006824.2
NP_006815.2
10969







(89)
(90)


EEF1A1
CCS3; EF1A; PTI1; CCS-
eukaryotic translation
An isoform of the
6q14.1
NM_001402.5
NP_001393.1
1915



3, EEF-1, EEF1A; EF-Tu;
elongation factor 1 alpha 1
alpha subunit of the

(91)
(92)



LENG7; eEF1A-1;

elongation factor-1



FLJ25721; GRAF-1EF;

complex, which is



MGC16224;

responsible for the



MGC102687;

enzymatic delivery of



MGC131894;

aminoacyl tRNAs to



HNGC: 16303

the ribosome.


EID3
NSE4B; NSMCE4B;
EP300 interacting inhibitor of
Unknown
12q23-q24.1
NM_001008394.1
NP_001008395.1
493861



FLJ25832
differentiation 3


(93)
(94)


ENO1
NNE; PPH; MPB1; MBP-
enolase 1, (alpha)
Encodes one of three
1p36.3-p36.2
NM_001428.2
NP_001419.1
2023



1; ENO1L1

enolase isoenzymes

(95)
(96)





found in mammals; it





encodes alpha-





enolase, a





homodimeric soluble





enzyme, and also





encodes a shorter





monomeric structural





lens protein, tau-





crystallin.


ERGIC1
ERGIC32; ERGIC-32;
endoplasmic reticulum-golgi
Encodes a cycling
5q35.1
NM_001031711.2
NP_001026881.1
57222



FLJ39864; KIAA1181;
intermediate compartment
membrane protein

(97)
(98)



MGC14345
(ERGIC) 1
which is an





endoplasmic





reticulum-golgi





intermediate





compartment





(ERGIC) protein





which interacts with





other members of this





protein family to





increase their





turnover.


FAM126B
HYCC2; FLJ10981;
family with sequence
Unknown
2q33.1
NM_173822.3
NP_776183.1
285172



FLJ37917; FLJ42947;
similarity 126, member B


(99)
(100)



MGC39518


FAM35A
FAM35A1; MGC5560;
family with sequence
Unknown
10q23.2
NM_019054.2
NP_061927.2
54537



bA163M19.1
similarity 35, member A


(101)
(102)


FAM55C
MST115; MSTP115;
family with sequence
Unknown
3q12.3
NM_001134456.1
NP_001127928.1
91775



FLJ30102; MGC15606
similarity 55, member C


(103)
(104)







NM_145037.2
NP_659474.1







(105)
(106)


FKBP2
PPIase; FKBP-13
FK506 binding protein 2,
Protein encoded by
11q13.1-q13.3
NM_001135208.1
NP_001128680.1
2286




13 kDa
this gene is a member

(107)
(108)





of the immunophilin

NM_004470.3
NP_004461.2





protein family, which

(109)
(110)





plays a role in

NM_057092.2
NP_476433.1





immunoregulation

(111)
(112)





and basic cellular





processes involving





protein folding and





trafficking.


FNDC3A
HUGO; FNDC3;
fibronectin type III domain
Unknown
13q14.2
NM_001079673.1
NP_001073141.1
22862



FLJ31509; KIAA0970;
containing 3A


(113)
(114)



bA203I16.1; bA203I16.5;



RP11-203I16.5


FNDC3B
FAD104; PRO4979;
fibronectin type III domain
Unknown
3q26.31
NM_001135095.1
NP_001128567.1
64778



FLJ23399; MGC10002;
containing 3B


(115)
(116)



YVTM2421;



NM_022763.3
NP_073600.3



DKFZp762K137;



(117)
(118)



DKFZp686D14170


G2E3
PHF7B; FLJ20333;
G2/M-phase specific E3
Unknown
14q12
NM_017769.3
NP_060239.2
55632



KIAA1333
ubiquitin ligase


(119)
(120)


GDF15
PDF; MIC1; PLAB; MIC-
growth differentiation factor
Member of the
19p13.11
NM_004864.2
NP_004855.2
9518



1; NAG-1; PTGFB; GDF-
15
transforming growth

(121)
(122)



15

factor-beta





superfamily and





regulate tissue





differentiation and





maintenance.


GEN1
Gen; FLJ40869;
Gen homolog 1, endonuclease
Unknown
2p24.2
NM_001130009.1
NP_001123481.1
348654



DKFZp781F0986



(123)
(124)







NM_182625.3
NP_872431.3







(125)
(126)


GLIS2
NPHP7; FLJ38247
GLIS family zinc finger 2
Encodes a nuclear
16p13.3
NM_032575.2
NP_115964.2
84662





transcription factor

(127)
(128)





with five C2H2-type





zinc finger domains.


GLUD1
GDH; GDH1; GLUD;
glutamate dehydrogenase 1
Gene encodes
10q23.3
NM_005271.3
NP_005262.1
2746



MGC132003

glutamate

(129)
(130)





dehydrogenase





protein; a





mitochondrial matrix





enzyme that catalyzes





the oxidative





deamination of





glutamate to alpha-





ketoglutarate and





ammonia.


GLYCTK
HBEBP2; HBEBP4;
glycerate kinase
Encoded enzyme
3p21.1
NM_001144951.1
NP_001138423.1
132158



HBeAgBP4A

catalyzes the

(131)
(132)





phosphorylation of

NM_145262.3
NP_660305.2





(R)-glycerate and

(133)
(134)





may be involved in





serine degradation





and fructose





metabolism.


GTF2H5
TTD; TFB5; TTDA;
general transcription factor
Encodes a subunit of
6q25.3
NM_207118.2
NP_997001.1
404672



TTD-A; TGF2H5;
IIH, polypeptide 5
transcription/repair

(135)
(136)



C6orf175; bA120J8.2

factor TFIIH, which





functions in gene





transcription and





DNA repair.


HAVCR1
KIM1; TIM1; HAVCR;
hepatitis A virus cellular
Protein encoded by
5q33.2
NM_001099414.1
NP_001092884.1
26762



KIM-1; TIM-1; TIMD1;
receptor 1
this gene is a

(137)
(138)



HAVCR-1

membrane receptor

NM_012206.2
NP_036338.2





for both human

(139)
(140)





hepatitis A virus and





TIMD4.


HLA-DMA
DMA; HLADM; RING6;
major histocompatibility
DM plays a central
6p21.3
NM_006120.3
NP_006111.2
3108



D6S222E
complex, class II, DM alpha
role in the peptide

(141)
(142)





loading of MHC class





II molecules by





helping to release the





CLIP molecule from





the peptide binding





site.


HNRNPH3
2H9; HNRPH3;
heterogeneous nuclear
Protein is involved in
10q22
NM_012207.2
NP_036339.1
3189



FLJ34092
ribonucleo-protein H3 (2H9)
the splicing process

(143)
(144)





and it also

NM_021644.3
NP_067676.2





participates in early

(145)
(146)





heat shock-induced





splicing arrest by





transiently leaving





the hnRNP





complexes.


HNRNPK
CSBP; TUNP; HNRPK;
heterogeneous nuclear
The protein encoded
9q21.32-q21.33
NM_002140.3
NP_002131.2
3190



FLJ41122
ribonucleo-protein K
by this gene is

(147)
(148)





located in the

NM_031262.2
NP_112552.1





nucleoplasm and has

(149)
(150)





three repeats of KH

NM_031263.2
NP_112553.1





domains that bind to

(151)
(152)





RNAs. It binds





tenaciously to





poly(C). This protein





is also thought to





have a role during





cell cycle progession.


HSP90AB4P
HSP90Bd; HsHsp90Bd
heat shock protein 90 kDa
Unknown
15q22.1
NR_002927.1

664618




alpha (cytosolic), class B


(153)




member 4 (pseudogene)


IARS2
FLJ10326
isoleucyl-tRNA synthetase 2,
Unknown
1q41
NM_018060.3
NP_060530.3
55699




mitochondrial


(154)
(155)


IL18
IGIF; IL-18; IL-1g;
interleukin 18 (interferon-
Protein encoded by
11q22.2-q22.3
NM_001562.2
NP_001553.1
3606



IL1F4; MGC12320
gamma-inducing factor)
this gene is a

(156)
(157)





proinflammatory





cytokine.


IL6ST
CD130; GP130; CDw130;
interleukin 6 signal
Protein functions as a
5q11
NM_002184.3
NP_002175.2
3572



IL6R-beta; GP130-RAPS
transducer (gp130, oncostatin
part of the cytokine

(158)
(159)




M receptor)
receptor complex.

NM_175767.1
NP_786943.1







(160)
(161)


ITGB1
CD29; FNRB; MDF2;
integrin, beta 1 (fibronectin
Involved in cell
10p11.2
NM_002211.3
NP_002202.2
3688



VLAB; GPIIA; MSK12;
receptor, beta polypeptide,
adhesion and

(162)
(163)



VLA-BETA
antigen CD29 includes
recognition in a

NM_033666.2
NP_389647.1




MDF2, MSK12)
variety of processes

(164)
(165)





including

NM_033667.2
NP_391987.1





embryogenesis,

(166)
(167)





hemostasis, tissue

NM_033668.2
NP_391988.1





repair, immune

(168)
(169)





response and

NM_033669.2
NP_391989.1





metastatic diffusion

(170)
(171)





of tumor cells.

NM_133376.2
NP_596867.1







(172)
(173)


KDM6B
JMJD3; KIAA0346
lysine (K)-specific
Unknown
17p13.1
NM_001080424.1
NP_001073893.1
23135




demethylase 6B


(174)
(175)


KAZALD1
BONO1; FKSG28;
Kazal-type serine peptidase
Gene encodes a
10q24.31
NM_030929.4
NP_112191.2
81621



FKSG40; FLJ24094;
inhibitor domain 1
secreted member of

(176)
(177)



IGFBP-rP10

the insulin growth





factor-binding protein





(IGFBP) superfamily,





may have a function





in bone development





and bone





regeneration.


KCTD1
C18orf5
potassium channel
Unknown
18q11.2
NM_001136205.1
NP_001129677.1
284252




tetramerisation domain


(178)
(179)




containing 1


NM_001142730.1
NP_001136202.1







(180)
(181)







NM_198991.2
NP_945342.1







(182)
(183)


KIAA1199
TMEM2L

Unknown
15q24
NM_018689.1
NP_061159.1
57214







(184)
(185)


KYNU

kynureninase (L-kynurenine
Involved in the
2q22.2
NM_001032998.1
NP_001028170.1
8942




hydrolase)
biosynthesis of NAD

(186)
(187)





cofactors from

NM_003937.2
NP_003928.1





tryptophan through

(188)
(189)





the kynurenine





pathway.


LMX1B
NPS1; LMX1.2;
LIM homeobox transcription
Functions as a
9q34
NM_002316.3
NP_002307.2
4010



MGC138325;
factor 1, beta
transcription factor,

(190)
(191)



MGC142051

and is essential for





the normal





development of





dorsal limb





structures, the





glomerular basement





membrane, the





anterior segment of





the eye, and





dopaminergic and





serotonergic neurons.


LNX2
PDZRN1; FLJ12933;
ligand of numb-protein X2
Unknown
13q12.2
NM_153371.3
NP_699202.1
222484



FLJ23932; FLJ38000;



(192)
(193)



MGC46315


MALAT1
HCN; NEAT2; MALAT-
metastasis associated lung
Unknown
11q13.1
NR_002819.2

378938



1; PRO2853;
adenocarcinoma transcript 1


(194)



NCRNA00047
(non-protein coding)


MAPK1IP1L
MISS; C14orf32;
mitogen-activated protein
Unknown
14q22.3
NM_144578.3
NP_653179.1
93487



MGC23138; c14_5346
kinase 1 interacting protein 1-


(195)
(196)




like


MAP4
MGC8617;
microtubule-associated
This protein
3p21
NM_001134364.1
NP_001127836.1
4134



DKFZp779A1753
protein 4
promotes microtubule

(197)
(198)





assembly, and has

NM_001134365.1
NP_001127837.1





been shown to

(199)
(200)





counteract

NM_002375.4
NP_002366.2





destabilization of

(201)
(202)





interphase

NM_030884.3
NP_112146.2





microtubule

(203)
(204)





catastrophe

NM_030885.3
NP_112147.2





promotion.

(205)
(206)


MGAT3
GNT3; GNT-III;
mannosyl (beta-1,4-)-
The enzyme encoded
22q13.1
NM_001098270.1
NP_001091740.1
4248



FLJ43371; MGC141943;
glycoprotein beta-1,4-N-
by this gene transfers

(207)
(208)



MGC142278
acetylglucosaminyltransferase
a GlcNAc residue to

NM_002409.4
NP_002400.3





the beta-linked

(209)
(210)





mannose of the





trimannosyl core of





N-linked





oligosaccharides and





produces a bisecting





GlcNAc.


MICALCL
Ebitein1; FLJ14966
MICAL C-terminal like
Unknown
11p15.3
NM_032867.2
NP_116256.2
84953







(211)
(212)


MIR194-1
MIRN194-1
microRNA 194-1
Involved in post-
1q41
NR_029711.1





transcriptional

(213)





regulation of gene





expression in





multicellular





organisms by





affecting both the





stability and





translation of





mRNAs.


MIR215
MIRN215; mir-215;
microRNA 215
Involved in post-
1q41
NR_029628.1

406997



miRNA215

transcriptional

(214)





regulation of gene





expression in





multicellular





organisms by





affecting both the





stability and





translation of





mRNAs.


MIR632
MIRN632; hsa-mir-632
microRNA 632
Involved in post-
17q11.2
NR_030362.1

693217





transcriptional

(215)





regulation of gene





expression in





multicellular





organisms by





affecting both the





stability and





translation of





mRNAs.


MIRLET7G
LET7G; MIRNLET7G;
microRNA let-7g
Involved in post-
3p21.1
NR_029660.1

406890



hsa-let-7g

transcriptional

(216)





regulation of gene





expression in





multicellular





organisms by





affecting both the





stability and





translation of





mRNAs.


MIRN15A
MIRN15A; miRNA15A;
microRNA 15a
Involved in post-
13q14.2
NR_029485.1

406948



hsa-mir-15a

transcriptional

(217)





regulation of gene





expression in





multicellular





organisms by





affecting both the





stability and





translation of





mRNAs.


MIRN16-1
MIRN16-1; miRNA16-1
microRNA 16-1
Involved in post-
13q14.2
NR_029486.1

406950





transcriptional

(218)





regulation of gene





expression in





multicellular





organisms by





affecting both the





stability and





translation of





mRNAs.


MIRN16-2
MIRN16-2; miRNA16-2
microRNA 16-2
Involved in post-
3q25.33
NR_029525.1

406951





transcriptional

(219)





regulation of gene





expression in





multicellular





organisms by





affecting both the





stability and





translation of





mRNAs.


MLF1IP
CENPU; KLIP1; PBIP1;
MLF1 interacting protein
An additional factor
4q35.1
NM_024629.3
NP_078905.2
79682



CENP-U; CENP50;

required for

(220)
(221)



CENP-50; FLJ23468;

centromere assembly.



CENP-U(50)


MLL5
KMT2E; FLJ10078;
myeloid/lymphoid or mixed-
Encodes a protein
7q22.1
NM_018682.3
NP_061152.3
55904



FLJ14026; HDCMC04P;
lineage leukemia 5 (trithorax
with an N-terminal

(222)
(223)



MGC70452
homolog, Drosophila)
PHD zinc finger and

NM_182931.2
NP_891847.1





a central SET

(224)
(225)





domain.





Overexpression of the





protein inhibits cell





cycle progression.


MRPL45
MGC11321
mitochondrial ribosomal
This gene encodes a
17q21.2
NM_032351.3
NP_115727.3
84311




protein L45
39S subunit protein

(226)
(227)





that helps in protein





synthesis within the





mitochondrion.


MRPS17P6

mitochondrial ribosomal
Unknown
14p11


359759




protein S17 pseudogene 6


MRPS18B
PTD017; S18amt;
mitochondrial ribosomal
This gene encodes a
6p21.3
NM_014046.3
NP_054765.1
28973



C6orf14; HSPC183;
protein S18B
28S subunit protein

(228)
(229)



MRPS18-2; HumanS18a;

that belongs to the



MRP-S18-2;

ribosomal protein



DKFZp564H0223

S18P family, and





helps in protein





synthesis within the





mitochondrion.


MTHFD1
MTHFC; MTHFD
methylenetetrahydrofolate
Gene encodes a
14q24
NM_005956.3
NP_005947.3
4522




dehydrogenase (NADP+
protein that possesses

(230)
(231)




dependent) 1,
three distinct




methenyltetrahydrofolate
enzymatic activities,




cyclohydrolase,
each of these




formyltetrahydrofolate
activities catalyzes




synthetase
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.


MTMR11
CRA; RP11-212K13.1
myotubularin related protein
Unknown
1q12-q21
NM_001145862.1
NP_001139334.1
10903




11


(232)
(233)







NM_181873.3
NP_870988.2







(234)
(235)


MYC
MRTL; c-Myc; bHLHe39
v-myc myelocytomatosis
Protein encoded by
8q24.21
NM_002467.4
NP_002458.2
4609




viral oncogene homolog
this gene is a

(236)
(237)




(avian)
multifunctional,

XM_001725300.1
XP_001725352.1





nuclear

(238)
(239)





phosphoprotein that

XM_001725281.1
XP_001725333.1





plays a role in cell

(240)
(241)





cycle progression,





apoptosis and cellular





transformation.


NANOGP4
NANOGP2
Nanog homeobox pseudogene 4
Unknown
7p15


414132


NAT13
SAN; MAK3; NAT5;
N-acetyltrans-ferase 13
Unknown
3q13.2
NM_025146.1
NP_079422.1
80218



hSAN; hNAT5;
(GCN5-related)


(242)
(243)



FLJ13194


NCRNA00099
UM9-5; UM9(5)
non-protein coding RNA 99
Unknown
4p15.2
NR_002813.1

359822







(244)


NDUFB3
B12
NADH dehydrogenase
Multisubunit
2q31.3
NM_002491.2
NP_002482.1
4709




(ubiquinone) 1 beta
NADH:ubiquinone

(245)
(246)




subcomplex, 3, 12 kDa
oxidoreductase





(complex I) is the





first enzyme complex





in the electron





transport chain of





mitochondria.


NDUFS5
Complex I-15 kDa; CI-15 kDa
NADH dehydrogenase
Encoded protein is a
1p34.2-p33
NM_004552.2
NP_004543.1
4725




(ubiquinone) Fe—S protein 5,
subunit of the

(247)
(248)




15 kDa (NADH-coenzyme Q
NADH:ubiquinone




reductase)
oxidoreductase





(complex I), the first





enzyme complex in





the electron transport





chain located in the





inner mitochondrial





membrane.


NFYA
HAP2; CBF-A; CBF-B;
nuclear transcription factor Y,
Protein encoded by
6p21.3
NM_002505.4
NP_002496.1
4800



NF-YA; FLJ11236
alpha
this gene is one

(249)
(250)





subunit of a trimeric

NM_021705.3
NP_068351.1





complex, forming a

(251)
(252)





highly conserved

XM_001718396.1
XP_001718448.1





transcription factor

(253)
(254)





that binds to CCAAT

XM_001716589.1
XP_001716641.1





motifs in the

(255)
(256)





promoter regions in a

XM_001717592.1
XP_001717644.1





variety of genes.

(257)
(258)


NPEPPS
PSA; MP100
aminopeptidase puromycin
Gene encodes the
17q21
NM_006310.3
NP_006301.3
9520




sensitive
puromycin-sensitive

(259)
(260)





aminopeptidase, a





zinc metallopeptidase





which hydrolyzes





amino acids from the





N-terminus of its





substrate.


NQO1
DTD; QR1; DHQU;
NAD(P)H dehydrogenase,
Gene is a member of
16q22.1
NM_000903.2
NP_000894.1
1728



DIA4; NMOR1; NMORI
quinone 1
the NAD(P)H

(261)
(262)





dehydrogenase

NM_001025433.1
NP_001020604.1





(quinone) family and

(263)
(264)





encodes a

NM_001025434.1
NP_001020605.1





cytoplasmic 2-

(265)
(266)





electron reductase.


NUAK2
SNARK; FLJ90349;
NUAK family, SNF1-like
Unknown
1q32.1
NM_030952.1
NP_112214.1
81788



DKFZp434J037;
kinase, 2


(267)
(268)



DKFZp686F01113


PAX8

paired box 8
This nuclear protein
2q12-q14
NM_003466.3
NP_003457.1
7849





is involved in thyroid

(269)
(270)





follicular cell

NM_013951.3
NP_039245.1





development and

(271)
(272)





expression of

NM_013952.3
NP_039246.1





thyroid-specific

(273)
(274)





genes.

NM_013953.3
NP_039247.1







(275)
(276)







NM_013992.3
NP_054698.1







(277)
(278)


PBLD
MAWBP; MAWDBP;
phenazine biosynthesis-like
Unknown
10pter-q25.3
NM_001033083.1
NP_001028255.1
64081



FLJ14767; FLJ35507
protein domain containing


(279)
(280)







NM_022129.3
NP_071412.2







(281)
(282)


PCBP2
HNRPE2; hnRNP-E2;
poly(rC) binding protein 2
Protein encoded by
12q13.12-q13.13
NM_001098620.2
NP_001092090.1
5094



MGC110998

this gene is one of the

(283)
(284)





major cellular

NM_001128911.1
NP_001122383.1





poly(rC)-binding

(285)
(286)





proteins. May be

NM_001128912.1
NP_001122384.1





involved in RNA

(287)
(288)





binding.

NM_001128913.1
NP_001122385.1







(289)
(290)







NM_001128914.1
NP_001122386.1







(291)
(292)







NM_005016.5
NP_005007.2







(293)
(294)







NM_031989.4
NP_114366.1







(295)
(296)


PDCD10
CCM3; TFAR15;
programmed cell death 10
Gene encodes an
3q26.1
NM_007217.3
NP_009148.2
11235



MGC1212; MGC24477

evolutionarily

(297)
(298)





conserved protein

NM_145859.1
NP_665858.1





associated with cell

(299)
(300)





apoptosis, the protein

NM_145860.1
NP_665859.1





interacts with the

(301)
(302)





serine/threonine





protein kinase MST4





to modulate the





extracellular signal-





regulated kinase





(ERK) pathway.


PKP1
B6P; MGC138829
plakophilin 1 (ectodermal
This protein may be
1q32
NM_000299.3
NP_000290.2
5317




dysplasia/skin fragility
involved in molecular

(303)
(304)




syndrome)
recruitment and

NM_001005337.1
NP_001005337.1





stabilization during

(305)
(306)





desmosome





formation.


PLCB3
FLJ37084
phospholipase C, beta 3
Catalyzes the
11q13
NM_000932.2
NP_000923.1
5331




(phosphatidylinositol-
production of the

(307)
(308)




specific)
secondary





messengers





diacylglycerol and





inositol 1,4,5-





triphosphate from





phosphatidylinositol





in G-protein-linked





receptor-mediated





signal transduction.


POLH
XPV; XP-V; RAD30;
polymerase (DNA directed),
Encodes a member of
6p21.1
NM_006502.2
NP_006493.1
5429



RAD30A; FLJ16395;
eta
the Y family of

(309)
(310)



FLJ21978

specialized DNA





polymerases, it





accurately replicates





UV-damaged DNA;





when thymine dimers





are present.


POU5F1P5

POU class 5 homeobox 1
Unknown
10q21.3


100009667




pseudogene 5


PPP1R10
FB19; CAT53; PNUTS;
protein phosphatase 1,
Gene encodes a
6p21.3
NM_002714.2
NP_002705.2
5514



PP1R10
regulatory (inhibitor) subunit
protein with

(311)
(312)




10
similarity to a rat





protein that has an





inhibitory effect on





protein phosphatase-1





(PP1).


PSIMCT-1

MCTS1 pseudogene
Unknown
20q11.2


100101490


PTRH2
BIT1; PTH2; CGI-147;
peptidyl-tRNA hydrolase 2
Unknown
17q23.1
NM_016077.3
NP_057161.1
51651



FLJ32471



(313)
(314)


PXN
FLJ16691
paxillin
Unknown
12q24.31
NM_001080855.1
NP_001074324.1
5829







(315)
(316)







NM_002859.2
NP_002850.2







(317)
(318)







NM_025157.3
NP_079433.3







(319)
(320)


QARS
GLNRS; PRO2195
glutaminyl-tRNA synthetase
Catalyzes the
3p21.3-p21.1
NM_005051.1
NP_005042.1
5859





aminoacylation of

(321)
(322)





tRNA by their





cognate amino acid.


RABL3
MGC23920
RAB, member of RAS
Unknown
3q13.33
NM_173825.3
NP_776186.2
285282




oncogene family-like 3


(323)
(324)


RAD51L1
REC2; R51H2; hREC2;
RAD51-like 1 (S. cerevisiae)
A suggested role of
14q23-q24.2
NM_002877.5
NP_002868.1
5890



RAD51B; MGC34245

this protein in sensing

(325)
(326)





DNA damage.

NM_133509.2
NP_598193.2







(327)
(328)







NM_133510.2
NP_598194.1







(329)
(330)


RBMX
RNMX; HNRPG;
RNA binding motif protein,
Gene belongs to the
Xq26.3
NM_002139.3
NP_002130.2
27316



RBMXP1; RBMXRT;
X-linked
RBMY gene family

(331)
(332)



hnRNP-G

which includes





candidate Y





chromosome





spermatogenesis





genes.


RGD1308059
MGC109234
similar to DNA segment, Chr
Unknown
5q31
NM_001025022.1
NP_001020193.1
362535




4, Brigham & Womens


(333)
(334)




Genetics 0951 expressed


RGD1309079
Ab2-095
similar to Ab2-095
Unknown
8q31
NM_001134472.1
NP_001127944.1
315891







(335)
(336)







XM_236493.4
XP_236493.4







(337)
(338)







XM_001068478.1
XP_001068478.1







(339)
(340)


RGD1309492

similar to mKIAA1737
Unknown
6q31
NM_001108044.2
NP_001101514.2
314330




protein


(341)
(342)







XM_234430.4
XP_234430.2







(343)
(344)







XM_001058624.1
XP_001058624.1







(345)
(346)


RMI1
BLAP75; C9orf76;
RecQ mediated genome
RMI1 is a component
9q21.32
NM_024945.2
NP_079221.2
80010



FLJ12888; RP11-346I8.1
instability 1, homolog (S. cerevisiae)
of protein complexes

(347)
(348)





that limit DNA





crossover formation





via the dissolution of





double Holliday





junctions.


RNU86
U82; U86
RNA, U86 small nucleolar
Involved in pre-
22q13
NR_000026.1

116936





rRNA processing and

(349)





modification.


RPL18AP3
bcm182;
ribosomal protein L18a
Unknown
12q23.3
NR_001593.1

390354



RPL18A_6_1273
pseudogene 3


(350)


RPL27A
FLJ43464; MGC10850;
ribosomal protein L27a
This gene encodes a
11p15
NM_000990.4
NP_000981.1
6157



MGC17878; MGC87238

ribosomal protein that

(351)
(352)





is a component of the





60S subunit.


RPL29P2
RPL29_10_1510
ribosomal protein L29
Unknown
17p13
NR_002778.1

118432




pseudogene 2


(353)


RPL29P31
RPL29_11_1549
ribosomal protein L29
Unknown
17q21.31
XM_210334.1
XP_210334.1
284064




pseudogene 31


(354)
(355)







XM_937685.1
XP_942778.1







(356)
(357)







XM_001722116.1
XP_001722168.1







(358)
(359)


RPL3
TARBP-B; MGC104284
ribosomal protein L3
This gene encodes a
22q13
NM_000967.3
NP_000958.1
6122





ribosomal protein that

(360)
(361)





is a component of the

NM_001033853.1
NP_001029025.1





60S subunit.

(362)
(363)


RPL34
MGC111005
ribosomal protein L34
This gene encodes a
4q25
NM_000995.3
NP_000986.2
6164





ribosomal protein that

(364)
(365)





is a component of the

NM_033625.2
NP_296374.1





60S subunit.

(366)
(367)


RPL37A
MGC74786
ribosomal protein L37a
This gene encodes a
2q35
NM_000998.4
NP_000989.1
6168





ribosomal protein that

(368)
(369)





is a component of the





60S subunit.


RPL4
RPL1
ribosomal protein L4
This gene encodes a
15q22
NM_000968.2
NP_000959.2
6124





ribosomal protein that

(370)
(371)





is a component of the





60S subunit.


RPL7A
TRUP; SURF3
ribosomal protein L7a
can interact with a
9q34
NM_000972.2
NP_000963.1
6130





subclass of nuclear

(372)
(373)





hormone receptors,





including thyroid





hormone receptor,





and inhibit their





ability to





transactivate by





preventing their





binding to their DNA





response elements.


RPS10P2
dJ858M22.1;
ribosomal protein S10
Unknown
20p12


140758



RPS10_15_1685
pseudogene 2


RPS18
KE3; HKE3; KE-3;
ribosomal protein S18
This gene encodes a
6p21.3
NM_022551.2
NP_072045.1
6222



D6S218E; MGC117351;

ribosomal protein that

(374)
(375)



MGC126835;

is a component of the



MGC126837

40S subunit.


RPS18P14
RPS18_10_1747
ribosomal protein S18
Unknown
22q12.2


100128535




pseudogene 14


RPS2P29
RPS2_9_695
ribosomal protein S2
Unknown
6p21.1


646294




pseudogene 29


RPS3P1
dJ631M13.3
ribosomal protein S3
Unknown
20p12


140754




pseudogene 1


RPSA
LRP; p40; 67LR; 37LRP;
ribosomal protein SA
This gene encodes a
3p22.2
NM_001012321.1
NP_001012321.1
3921



LAMBR; LAMR1

high-affinity, non-

(376)
(377)





integrin family,

NM_002295.4
NP_002286.2





laminin receptor 1.

(378)
(379)


SCYE1P
bA400P21.1
small inducible cytokine
Unknown
20p12.1


170547




subfamily E, member 1




(endothelial monocyte-




activating) pseudogene


SDF2
SDF-2
stromal cell-derived factor 2
Protein encoded by
17q11.2
NM_006923.2
NP_008854.2
6388





this gene is believed

(380)
(381)





to be a secretory





protein.


SEMA3C
SemE; SEMAE
sema domain,
Unknown
7q21-q31
NM_006379.2
NP_006370.1
10512




immunoglobulin domain (Ig),


(382)
(383)




short basic domain, secreted,




(semaphorin) 3C


SERAC1
FLJ14917; FLJ30544
serine active site containing 1
Unknown
6q25.3
NM_032861.3
NP_116250.3
84947







(384)
(385)


SERPINI1
PI12; neuroserpin;
serpin peptidase inhibitor,
Protein is primarily
3q26.1
NM_001122752.1
NP_001116224.1
5274



DKFZp781N13156
clade I (neuroserpin), member 1
secreted by axons in

(386)
(387)





the brain, and

NM_005025.4
NP_005016.1





preferentially reacts

(388)
(389)





with and inhibits





tissue-type





plasminogen





activator.


SF3B4
SAP49; SF3b49;
splicing factor 3b, subunit 4,
The protein encoded
1q12-q21
NM_005850.3
NP_005841.1
10262



MGC10828
49 kDa
by this gene cross-

(390)
(391)





links to a region in





the pre-mRNA





immediately





upstream of the





branchpoint sequence





in pre-mRNA in the





prespliceosomal





complex A.


SFRS3
SRp20
splicing factor,
Unknown
6p21
NM_003017.4
NP_003008.1
6428




arginine/serine-rich 3


(392)
(393)


SFXN1
FLJ12876
sideroflexin 1
Unknown
5q35.2
NM_022754.5
NP_073591.2
94081







(394)
(395)


SKIL
SNO; SnoA; SnoI; SnoN
SKI-like oncogene
Unknown
3q26
NM_001145097.1
NP_001138569.1
6498







(396)
(397)







NM_001145098.1
NP_001138570.1







(398)
(399)







NM_005414.3
NP_005405.2







(400)
(401)


SLC25A25
MCSC; PCSCL;
solute carrier family 25
Unknown
9q34.11
NM_001006641.1
NP_001006642.1
114789



SCAMC-2; KIAA1896;
(mitochondrial carrier;


(402)
(403)



MGC105138;
phosphate carrier), member


NM_001006642.1
NP_001006643.1



MGC119514;
25


(404)
(405)



MGC119515;



NM_001006643.1
NP_001006644.1



MGC119516;



(406)
(407)



MGC119517; RP11-



NM_052901.2
NP_443133.2



395P17.4



(408)
(409)


SLC38A2
ATA2; SAT2; SNAT2;
solute carrier family 38,
Unknown
12q
NM_018976.4
NP_061849.2
54407



PRO1068; KIAA1382
member 2


(410)
(411)


SLC39A14
ZIP14; cig19; LZT-Hs4;
solute carrier family 39 (zinc
SLC39A14 belongs
8p21.3
NM_001128431.2
NP_001121903.1
23516



KIAA0062
transporter), member 14
to a subfamily of

(412)
(413)





proteins that show

NM_001135153.1
NP_001128625.1





structural

(414)
(415)





characteristics of zinc

NM_001135154.1
NP_001128626.1





transporters.

(416)
(417)







NM_015359.4
NP_056174.2







(418)
(419)


SMC6
SMC6L1; FLJ22116;
structural maintenance of
Unknown
2p24.2
NM_001142286.1
NP_001135758.1
79677



FLJ35534
chromosomes 6


(420)
(421)







NM_024624.5
NP_078900.1







(422)
(423)


SNORA1
ACA1
small nucleolar RNA,
Unknown
11q21
NR_003026.1

677792




H/ACA box 1


(424)


SNORA18
ACA18
small nucleolar RNA,
Unknown
11q21
NR_002959.1

677805




H/ACA box 18


(425)


SNORA19
ACA19
small nucleolar RNA,
Unknown
10q26
NR_002917.1

641451




H/ACA box 19


(426)


SNORA3
ACA3
small nucleolar RNA,
Unknown
11p15
NR_002580.1

619562




H/ACA box 3


(427)


SNORA32
ACA32
small nucleolar RNA,
Unknown
11q21
NR_003032.1

692063




H/ACA box 32


(428)


SNORA38
ACA38
small nucleolar RNA,
Unknown
6p21.33
NR_002971.1

677820




H/ACA box 38


(429)


SNORA40
ACA40
small nucleolar RNA,
Unknown
11q21
NR_002973.1

677822




H/ACA box 40


(430)


SNORA45
ACA3-2
small nucleolar RNA,
Involved in RNA
11p15.4
NR_002977.1

677826




H/ACA box 45
processing.

(431)


SNORA54
ACA54
small nucleolar RNA,
Unknown
11p15.4
NR_002982.1

677833




H/ACA box 54


(432)


SNORA6
ACA6
small nucleolar RNA,
Unknown
3p22.2
NR_002325.1

574040




H/ACA box 6


(433)


SNORA62
E2; E2-1; RNE2;
small nucleolar RNA,
Unknown
3p22
NR_002324.1

6044



RNU108
H/ACA box 62


(434)


SNORA76
ACA62
small nucleolar RNA,
Unknown
17q23.3
NR_002995.1

677842




H/ACA box 76


(435)


SNORA8
ACA8
small nucleolar RNA,
Unknown
11q21
NR_002920.1

654320




H/ACA box 8


(436)


SNORA84

small nucleolar RNA,
Unknown
9q22.31
NR_003704.2

100124534




H/ACA box 84


(437)


SNORD16
U16
small nucleolar RNA, C/D
Unknown
15q22
NR_002440.1

595097




box 16


(438)


SNORD18A
U18A
small nucleolar RNA, C/D
Unknown
15q22
NR_002441.1

595098




box 18A


(439)


SNORD18B
U18B
small nucleolar RNA, C/D
Unknown
15q22
NR_002442.1

595099




box 18B


(440)


SNORD18C
U18C
small nucleolar RNA, C/D
Unknown
15q22
NR_002443.1

595100




box 18C


(441)


SNORD24
U24; RNU24
small nucleolar RNA, C/D
Unknown
9q34
NR_002447.1

26820




box 24


(442)


SNORD35B
U35B; RNU35B
small nucleolar RNA, C/D
Unknown
19q13.3
NR_001285.1

84546




box 35B


(443)


SNORD36A
U36a; RNU36A
small nucleolar RNA, C/D
Unknown
9q34
NR_002448.1

26815




box 36A


(444)


SNORD36B
U36b; RNU36B
small nucleolar RNA, C/D
Unknown
9q34
NR_000017.1

26814




box 36B


(445)


SNORD36C
U36c; RNU36C
small nucleolar RNA, C/D
Unknown
9q34
NR_000016.1

26813




box 36C


(446)


SNORD3B-2
U3b2; U3-2B
small nucleolar RNA, C/D
Unknown
17p11.2
NR_003924.1

780852




box 3B-2


(447)


SNORD43
U43; RNU43
small nucleolar RNA, C/D
Involved in pre-
22q13
NR_002439.1

26807




box 43
rRNA processing and

(448)





modification.


SNORD44
U44; RNU44
small nucleolar RNA, C/D
Unknown
1q25.1
NR_002750.2

26806




box 44


(449)


SNORD47
U47; RNU47
small nucleolar RNA, C/D
Unknown
1q25.1
NR_002746.1

26802




box 47


(450)


SNORD5
mgh28S-2410
small nucleolar RNA, C/D
Unknown
11q21
NR_003033.1

692072




box 5


(451)


SNORD58A
U58a; RNU58A
small nucleolar RNA, C/D
Unknown
18q21
NR_002571.1

26791




box 58A


(452)


SNORD6
mgh28S-2412
small nucleolar RNA, C/D
Unknown
11q21
NR_003036.1

692075




box 6


(453)


SNORD60
U60; RNU60
small nucleolar RNA, C/D
Unknown
16p13.3
NR_002736.1

26788




box 60


(454)


SNORD61
U61; RNU61
small nucleolar RNA, C/D
Involved in RNA
Xq26.3
NR_002735.1

26787




box 61
processing.

(455)


SNORD74
U74; Z18
small nucleolar RNA, C/D
Unknown
1q25.1
NR_002579.1

619498




box 74


(456)


SNORD75
U75
small nucleolar RNA, C/D
Unknown
1q25.1
NR_003941.1

692195




box 75


(457)


SNORD76
U76
small nucleolar RNA, C/D
Unknown
1q25.1
NR_003942.1

692196




box 76


(458)


SNORD77
U77
small nucleolar RNA, C/D
Unknown
1q25.1
NR_003943.1

692197




box 77


(459)


SNORD78
U78
small nucleolar RNA, C/D
Unknown
1q25.1
NR_003944.1

692198




box 78


(460)


SNORD80
U80; Z15
small nucleolar RNA, C/D
Unknown
1q25.1
NR_003940.1

26774




box 80


(461)


SNORD81
U81; Z23
small nucleolar RNA, C/D
Unknown
1q25.1
NR_003938.1

26769




box 81


(462)


SNORD83A
U83A; RNU83A
small nucleolar RNA, C/D
Involved in pre-
22q13
NR_000027.1

116937




box 83A
rRNA processing and

(463)





modification.


SNORD83B
U83B; RNU83B
small nucleolar RNA, C/D
Involved in pre-
22q13
NR_000028.1

116938




box 83B
rRNA processing and

(464)





modification.


SPATA19
SPAS1; FLJ25851;
spermato-genesis associated
Unknown
11q25
NM_174927.1
NP_777587.1
219938



spergen1
19


(465)
(466)


SRP54

signal recognition particle
Unknown
14q13.2
NM_001146282.1
NP_001139754.1
6729




54 kDa


(467)
(468)







NM_003136.3
NP_003127.1







(469)
(470)


ST6GAL2
SIAT2; FLJ30711;
ST6 beta-galactosamide
Catalyzes the transfer
2q11.2-q12.1
NM_001142351.1
NP_001135823.1
84620



FLJ37730; FLJ38334;
alpha-2,6-sialyltranferase 2
of sialic acid from

(471)
(472)



KIAA1877; ST6GalII

CMP-sialic acid to an

NM_001142352.1
NP_001135824.1





acceptor

(473)
(474)





carbohydrate, usually

NM_032528.2
NP_115917.1





to the terminal ends

(475)
(476)





of carbohydrate





chains.


STH
MAPTIT; MGC163191;
saitohin
Unknown
17q21.1
NM_001007532.2
NP_001007533.1
246744



MGC163193



(477)
(478)


SUPT6H
SPT6; SPT6H; emb-5;
suppressor of Ty 6 homolog
Unknown
17q11.2
NM_003170.3
NP_003161.2
6830



KIAA0162; MGC87943
(S. cerevisiae)


(479)
(480)


TAF1
OF; BA2R; CCG1;
TAF1 RNA polymerase II,
This gene encodes
Xq13.1
NM_004606.3
NP_004597.2
6872



CCGS; DYT3; KAT4;
TATA box binding protein
the largest subunit of

(481)
(482)



P250; NSCL2; TAF2A;
(TBP)-associated factor
TFIID. This subunit

NM_138923.2
NP_620278.1



N-TAF1; TAFII250

binds to core

(483)
(484)





promoter sequences





encompassing the





transcription start





site.


TAF1D
JOSD3; MGC5306;
TATA box binding protein
Plays a role in RNA
11q21
NM_024116.3
NP_077021.1
79101



TAF(I)41
(TBP)-associated factor,
polymerase I

(485)
(486)




RNA polymerase I, D, 41 kDa
transcription.


TBC1D5
KIAA0210
TBC1 domain family,
Unknown
3p24.3
NM_001134380.1
NP_001127852.1
9779




member 5


(487)
(488)







NM_001134381.1
NP_001127853.1







(489)
(490)







NM_014744.2
NP_055559.1







(491)
(492)


TEX2
HT008; TMEM96;
testis expressed 2
Unknown
17q23.3
NM_018469.3
NP_060939.3
55852



KIAA1738;



(493)
(494)



DKFZp781G0721


TEX21
MGC114468; Tex21
testis expressed gene 21
Unknown
6q24
NM_001025658.1
NP_001020829.1
299152







(495)
(496)


TMEM49
VMP1; DKFZp566I133
transmembrane protein 49
Unknown
17q23.1
NM_030938.3
NP_112200.2
81671







(497)
(498)


TNPO1
MIP; TRN; IPO2; MIP1;
transportin 1
Interacts with nuclear
5q13.2
NM_002270.3
NP_002261.3
3842



KPNB2

localization signals to

(499)
(500)





target nuclear

NM_153188.2
NP_694858.1





proteins to the

(501)
(502)





nucleus.


TRAF7
RFWD1; RNF119;
TNF receptor-associated
Signal transducer for
16p13.3
NM_032271.2
NP_115647.2
84231



MGC7807;
factor 7
members of the TNF

(503)
(504)



DKFZp586I021

receptor superfamily.


TRIM66
TIF1D; C11orf29;
tripartite motif-containing 66
Unknown
11p15.4
NM_014818.1
NP_055633.1
9866



FLJ10046; TIF1DELTA



(505)
(506)







XM_001716253.1
XP_001716305.1







(507)
(508)







XM_001716830.1
XP_001716882.1







(509)
(510)







XM_001717903.1
XP_001717955.1







(511)
(512)


TSGA13

testis specific, 13
Unknown
7q32
NM_052933.2
NP_443165.1
114960







(513)
(514)


TUBD1
TUBD; FLJ12709
tubulin, delta 1
Unknown
17q23.1
NM_016261.2
NP_057345.2
51174







(515)
(516)


TYW3
C1orf171; FLJ40918
tRNA-yW synthesizing
YW3 is the human
1p31.1
NM_138467.2
NP_612476.1
127253




protein 3 homolog (S. cerevisiae)
homolog of a yeast

(517)
(518)





gene essential for yW





synthesis.


U58

U58 small nucleolar RNA
Unknown
18q21
NR_002573.1

619491







(519)


UBA52
CEP52; RPL40;
ubiquitin A-52 residue
This gene encodes a
19p13.1-p12
NM_001033930.1
NP_001029102.1
7311



HUBCEP52; MGC57125;
ribosomal protein fusion
fusion protein

(520)
(521)



MGC126879;
product 1
consisting of

NM_003333.3
NP_003324.1



MGC126881

ubiquitin at the N

(522)
(523)





terminus and





ribosomal protein





L40 at the C





terminus, a C-





terminal extension





protein (CEP).


USP10
UBPO; MGC2621;
ubiquitin specific peptidase
The enzyme
16q24.1
NM_005153.2
NP_005144.2
9100



KIAA0190
10
specifically cleaves

(524)
(525)





ubiquitin from





ubiquitin-conjugated





protein substrates.


VOF16
Vof-16; Vof16
ischemia related factor vof-16
Displays increased
8q22
NM_147207.1
NP_671740.1
259227





mRNA expression in

(526)
(527)





permanent ischemic





brain


WDR51B
TUWD12; FLJ14923;
WD repeat domain 51B
Unknown
12q21.33
NM_172240.1
NP_758440.1
282809



FLJ41111



(528)
(529)


WDR81
FLJ23776; FLJ33817
WD repeat domain 81
Unknown
17p13.3
NM_152348.2
NP_689561.2
124997







(530)
(531)







NM_001163809.1
NP_001157281.1







(532)
(533)







NM_001163811.1
NP_001157283.1







(534)
(535)







NM_152348.3
NP_689561.2







(536)
(537)


WDR82
SWD2; MST107;
WD repeat domain 82
A component of the
3p21.2-p21.1
NM_025222.3
NP_079498.2
80335



WDR82A; MSTP107;

mammalian SET1A

(538)
(539)



PRO2730; TMEM113;

(MIM



PRO34047

611052)/SET1B





(MIM 611055)





histone H3-Lys4





methyltrans-ferase





complexes.


WIPF2
WICH; WIRE
WAS/WASL interacting
This protein has a
17q21.1-q21.2
NM_133264.4
NP_573571.1
147179




protein family, member 2
role in the WASP-

(540)
(541)





mediated





organization of the





actin cytoskeleton





and that this protein





is a potential link





between the activated





platelet-derived





growth factor





receptor and the actin





polymerization





machinery.


ZBTB37
MGC2629;
zinc finger and BTB domain
Unknown
1q25.1
NM_001122770.1
NP_001116242.1
84614



D430004I08Rik
containing 37


(542)
(543)







NM_032522.3
NP_115911.1







(544)
(545)


ZC3H4
C19orf7; KIAA1064
zinc finger CCCH-type
Unknown
19q13.32
NM_015168.1
NP_055983.1
23211




containing 4


(546)
(547)


ZHX2
RAF; AFR1; KIAA0854
zinc fingers and homeoboxes 2
Nuclear homodimeric
8q24.13
NM_014943.3
NP_055758.1
22882





transcriptional

(548)
(549)





repressor that





interacts with the A





subunit of nuclear





factor-Y (NF-YA)





and contains two





C2H2-type zinc





fingers and five





homeobox DNA-





binding domains.


ZMYND8
RACK7; PRKCBP1;
zinc finger, MYND-type
Protein encoded by
20q13.12
NM_012408.3
NP_036540.3
23613



PRO2893; MGC31836
containing 8
this gene is a receptor

(550)
(551)





for activated C-kinase

NM_183047.1
NP_898868.1





(RACK) protein.

(552)
(553)







NM_183048.1
NP_898869.1







(554)
(555)


ZNF143
SBF; STAF; pHZ-1
zinc finger protein 143
Unknown
11p15.4
NM_003442.5
NP_003433.3
7702







(556)
(557)


ZNF662
FLJ33347; FLJ45880;
zinc finger protein 662
Unknown
3p22.1
NM_001134656.1
NP_001128128.1
389114



MGC149141



(558)
(559)







NM_207404.3
NP_997287.2







(560)
(561)


ZWILCH
KNTC1AP; hZwilch;
Zwilch, kinetochore
Unknown
15q22.31
NM_017975.3
NP_060445.3
55055



FLJ10036; FLJ16343;
associated, homolog


(562)
(563)



MGC111034
(Drosophila)









As used herein, the term “nucleic acid” refers to single or multiple stranded molecules, which may be DNA or RNA, or any combination thereof, including modifications to those nucleic acids. The nucleic acid may represent a coding strand or its complement, or any combination thereof. Nucleic acids may be identical in sequence to the sequences which are naturally occurring for any of the moieties discussed herein or may include alternative codons which encode the same amino acid as that which is found in the naturally occurring sequence. These nucleic acids can also be modified from their typical structure. Such modifications include, but are not limited to, methylated nucleic acids, the substitution of a non-bridging oxygen on the phosphate residue with either a sulfur (yielding phosphorothioate deoxynucleotides), selenium (yielding phosphorselenoate deoxynucleotides), or methyl groups (yielding methylphosphonate deoxynucleotides), a reduction in the AT content of AT rich regions, or replacement of non-preferred codon usage of the expression system to preferred codon usage of the expression system. The nucleic acid can be directly cloned into an appropriate vector, or if desired, can be modified to facilitate the subsequent cloning steps. Such modification steps are routine, an example of which is the addition of oligonucleotide linkers which contain restriction sites to the termini of the nucleic acid. General methods are set forth in Sambrook et al. (2001) Molecular Cloning—A Laboratory Manual (3rd ed.) Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor Press, NY, (Sambrook).


Once the nucleic acid sequence is obtained, the sequence encoding the specific amino acids can be modified or changed at any particular amino acid position by techniques well known in the art. For example, PCR primers can be designed which span the amino acid position or positions and which can substitute any amino acid for another amino acid. Alternatively, one skilled in the art can introduce specific mutations at any point in a particular nucleic acid sequence through techniques for point mutagenesis. General methods are set forth in Smith, M. “In vitro mutagenesis” Ann. Rev. Gen., 19:423-462 (1985) and Zoller, M. J. “New molecular biology methods for protein engineering” Curr. Opin. Struct. Biol., 1:605-610 (1991), which are incorporated herein in their entirety for the methods. These techniques can be used to alter the coding sequence without altering the amino acid sequence that is encoded.


The sequences contemplated herein include full-length wild-type (or native) sequences, as well as allelic variants, variants, fragments, homologs or fusion sequences that retain the ability to function as the cellular nucleic acid or protein involved in viral infection. In certain examples, a protein or nucleic acid sequence has at least 50% sequence identity, for example at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 98% sequence identity to a native sequences of the genes set forth in Table 1. In other examples, a nucleic acid sequence involved in viral infection has a sequence that hybridizes to a sequence of a gene set forth in Table 1 and retains the activity of the sequence of the gene set forth in Table 1. For example, and not to be limiting, a nucleic acid that hybridizes to an AHR nucleic acid sequence and encodes a protein that retains AHR activity is contemplated by the present invention. Such sequences include the genomic sequence for the genes set forth in Table 1. The examples set forth above for AHR are merely illustrative and should not be limited to AHR as the analysis set forth in this example applies to every nucleic acid and protein for the genes listed in Table 1.


Unless otherwise specified, any reference to a nucleic acid molecule includes the reverse complement of the nucleic acid. For example, any siRNA sequence set forth herein can also include the reverse complement of that sequence. Except where single-strandedness is required by the text herein (for example, a ssRNA molecule), any nucleic acid written to depict only a single strand encompasses both strands of a corresponding double-stranded nucleic acid. For example, depiction of a plus-strand of a dsDNA also encompasses the complementary minus-strand of that dsDNA. Additionally, and reference to the nucleic acid molecule that encodes a specific protein, or a fragment thereof, encompasses both the sense strand and its relevant complement. Fragments of the nucleic acids for the genes set forth in Table 1 and throughout the specification are also contemplated. These fragments can be utilized as primers and probes to amplify, inhibit or detect any of the nucleic acids or genes set forth in Table 1.


Hybridization conditions resulting in particular degrees of stringency will vary depending upon the nature of the hybridization method and the composition and length of the hybridizing nucleic acid sequences. Generally, the temperature of hybridization and the ionic strength (such as the Na+ concentration) of the hybridization buffer will determine the stringency of hybridization. Calculations regarding hybridization conditions for attaining particular degrees of stringency are discussed in Sambrook et al., (1989) Molecular Cloning, second edition, Cold Spring Harbor Laboratory, Plainview, N.Y. (chapters 9 and 11). The following is an exemplary set of hybridization conditions and is not limiting:


Very High Stringency (Detects Sequences that Share 90% Identity)


Hybridization: 5×SSC at 65° C. for 16 hours


Wash twice: 2×SSC at room temperature (RT) for 15 minutes each


Wash twice: 0.5×SSC at 65° C. for 20 minutes each


High Stringency (Detects Sequences that Share 80% Identity or Greater)


Hybridization: 5×-6×SSC at 65° C.-70° C. for 16-20 hours


Wash twice: 2×SSC at RT for 5-20 minutes each


Wash twice: 1×SSC at 55° C.-70° C. for 30 minutes each


Low Stringency (Detects Sequences that Share Greater than 50% Identity)


Hybridization: 6×SSC at RT to 55° C. for 16-20 hours


Wash at least twice: 2×-3×SSC at RT to 55° C. for 20-30 minutes each.


Also provided is a vector, comprising a nucleic acid set forth herein. The vector can direct the in vivo or in vitro synthesis of any of the proteins or polypeptides described herein. The vector is contemplated to have the necessary functional elements that direct and regulate transcription of the inserted nucleic acid. These functional elements include, but are not limited to, a promoter, regions upstream or downstream of the promoter, such as enhancers that may regulate the transcriptional activity of the promoter, an origin of replication, appropriate restriction sites to facilitate cloning of inserts adjacent to the promoter, antibiotic resistance genes or other markers which can serve to select for cells containing the vector or the vector containing the insert, RNA splice junctions, a transcription termination region, or any other region which may serve to facilitate the expression of the inserted gene or hybrid gene (See generally, Sambrook et al.). The vector, for example, can be a plasmid. The vectors can contain genes conferring hygromycin resistance, ampicillin resistance, gentamicin resistance, neomycin resistance or other genes or phenotypes suitable for use as selectable markers, or methotrexate resistance for gene amplification.


There are numerous other E. coli (Escherichia coli) expression vectors, known to one of ordinary skill in the art, which are useful for the expression of the nucleic acid insert. Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species. In these prokaryotic hosts one can also make expression vectors, which will typically contain expression control sequences compatible with the host cell (e.g., an origin of replication). In addition, any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (Trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda. Additionally, yeast expression can be used. The invention provides a nucleic acid encoding a polypeptide of the present invention, wherein a yeast cell can express the nucleic acid. More specifically, the nucleic acid can be expressed by Pichia pastoris or S. cerevisiae.


Mammalian cells also permit the expression of proteins in an environment that favors important post-translational modifications such as folding and cysteine pairing, addition of complex carbohydrate structures, and secretion of active protein. Vectors useful for the expression of active proteins are known in the art and can contain genes conferring hygromycin resistance, genticin or G418 resistance, or other genes or phenotypes suitable for use as selectable markers, or methotrexate resistance for gene amplification. A number of suitable host cell lines capable of secreting intact human proteins have been developed in the art, and include the CHO cell lines, HeLa cells, COS-7 cells, myeloma cell lines, Jurkat cells, etc. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer, and necessary information processing sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Preferred expression control sequences are promoters derived from immunoglobulin genes, SV40, Adenovirus, Bovine Papilloma Virus, etc.


The expression vectors described herein can also include nucleic acids of the present invention under the control of an inducible promoter such as the tetracycline inducible promoter or a glucocorticoid inducible promoter. The nucleic acids of the present invention can also be under the control of a tissue-specific promoter to promote expression of the nucleic acid in specific cells, tissues or organs. Any regulatable promoter, such as a metallothionein promoter, a heat-shock promoter, and other regulatable promoters, of which many examples are well known in the art are also contemplated. Furthermore, a Cre-loxP inducible system can also be used, as well as an Flp recombinase inducible promoter system, both of which are known in the art.


Insect cells also permit the expression of mammalian proteins. Recombinant proteins produced in insect cells with baculovirus vectors undergo post-translational modifications similar to that of wild-type proteins. The invention also provides for the vectors containing the contemplated nucleic acids in a host suitable for expressing the nucleic acids. The host cell can be a prokaryotic cell, including, for example, a bacterial cell. More particularly, the bacterial cell can be an E. coli cell. Alternatively, the cell can be a eukaryotic cell, including, for example, a Chinese hamster ovary (CHO) cell, a COS-7 cell, a HELA cell, an avian cell, a myeloma cell, a Pichia cell, or an insect cell. A number of other suitable host cell lines have been developed and include myeloma cell lines, fibroblast cell lines, a cell line suitable for infection by a pathogen, and a variety of tumor cell lines such as melanoma cell lines. The vectors containing the nucleic acid segments of interest can be transferred into the host cell by well-known methods, which vary depending on the type of cellular host. For example, calcium chloride transformation is commonly utilized for prokaryotic cells, whereas calcium phosphate, DEAE dextran, Lipofectamine, or lipofectin mediated transfection, electroporation or any method now known or identified in the future can be used for other eukaryotic cellular hosts.


Polypeptides

The present invention provides isolated polypeptides comprising the polypeptide or protein sequences set forth under the GenBank Accession Nos. set forth in Table 1. The present invention also provides fragments of these polypeptides. These fragments can be of sufficient length to serve as antigenic peptides for the generation of antibodies. The present invention also contemplates functional fragments that possess at least one activity of a gene or gene product listed in Table 1, for example, involved in viral infection. It will be known to one of skill in the art that each of the proteins set forth herein possess other properties, such as for example, AHR is an aromate hydrocarbon receptor, and AK5 has adenylate kinase activity. Fragments and variants of the proteins set forth herein can include one or more conservative amino acid residues as compared to the amino acid sequence listed under their respective GenBank Accession Nos.


By “isolated polypeptide” or “purified polypeptide” is meant a polypeptide that is substantially free from the materials with which the polypeptide is normally associated in nature or in culture. The polypeptides of the invention can be obtained, for example, by extraction from a natural source if available (for example, a mammalian cell), by expression of a recombinant nucleic acid encoding the polypeptide (for example, in a cell or in a cell-free translation system), or by chemically synthesizing the polypeptide. In addition, a polypeptide can be obtained by cleaving full-length polypeptides. When the polypeptide is a fragment of a larger naturally occurring polypeptide, the isolated polypeptide is shorter than and excludes the full-length, naturally occurring polypeptide of which it is a fragment.


Also provided by the present invention is a polypeptide comprising an amino acid sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the polypeptide sequences set forth under the GenBank Accession Nos. found in Table 1.


It is understood that as discussed herein the use of the terms “homology” and “identity” mean the same thing as similarity. Thus, for example, if the use of the word homology is used to refer to two non-natural sequences, it is understood that this is not necessarily indicating an evolutionary relationship between these two sequences, but rather is looking at the similarity or relatedness between their nucleic acid sequences. Many of the methods for determining homology between two evolutionarily related molecules are routinely applied to any two or more nucleic acids or proteins for the purpose of measuring sequence similarity regardless of whether they are evolutionarily related.


In general, it is understood that one way to define any known variants and derivatives or those that might arise, of the disclosed nucleic acids and polypeptides herein, is through defining the variants and derivatives in terms of homology to specific known sequences. In general, variants of nucleic acids and polypeptides herein disclosed typically have at least, about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent homology to the stated sequence or the native sequence. Those of skill in the art readily understand how to determine the homology of two or more polypeptides or nucleic acids. For example, the homology can be calculated after aligning the two sequences so that the homology is at its highest level.


Another way of calculating homology can be performed by published algorithms. Optimal alignment of sequences for comparison can be conducted by the local homology algorithm of Smith and Waterman Adv. Appl. Math. 2: 482 (1981), by the homology alignment algorithm of Needleman and Wunsch, J. Mol. Biol. 48: 443 (1970), by the search for similarity method of Pearson and Lipman, Proc. Natl. Acad. Sci. U.S.A. 85: 2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.; the BLAST algorithm of Tatusova and Madden FEMS Microbiol. Lett. 174: 247-250 (1999) available from the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/blast/bl2seq/bl2.html), or by inspection.


The same types of homology can be obtained for nucleic acids by for example the algorithms disclosed in Zuker, M. Science 244:48-52, 1989, Jaeger et al. Proc. Natl. Acad. Sci. USA 86:7706-7710, 1989, Jaeger et al. Methods Enzymol. 183:281-306, 1989, which are herein incorporated by, reference for at least material related to nucleic acid alignment. It is understood that any of the methods typically can be used and that in certain instances the results of these various methods may differ, but the skilled artisan understands if identity is found with at least one of these methods, the sequences would be said to have the stated identity.


For example, as used herein, a sequence recited as having a particular percent homology to another sequence refers to sequences that have the recited homology as calculated by any one or more of the calculation methods described above. For example, a first sequence has 80 percent homology, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent homology to the second sequence using the Zuker calculation method even if the first sequence does not have 80 percent homology to the second sequence as calculated by any of the other calculation methods. As yet another example, a first sequence has 80 percent homology, as defined herein, to a second sequence if the first sequence is calculated to have 80 percent homology to the second sequence using each of calculation methods (although, in practice, the different calculation methods will often result in different calculated homology percentages).


Also provided by the present invention are polypeptides set forth under the GenBank Accession Nos. disclosed herein, or fragments thereof, with one or more conservative amino acid substitutions. These conservative substitutions are such that an amino acid having similar properties replaces a naturally occurring one. Such conservative substitutions do not alter the function of the polypeptide. For example, conservative substitutions can be made as follows: Arg can be replaced with Lys, Asn can be replace with Gln, Asn can be replaced with Glu, Cys can be replaced with Ser, Gln can be replaced with Asn, Glu can be replaced with Asp, Gly can be replaced with Pro, His can be replaced with Gln, Ile can be replaced with Leu or Val, Gly can be replaced with Pro, His can be replaced with Gln, Ile can be replaced with Ile or Val, Leu can be replaced with Ile or Val, Lys can be replaced with Arg or Gln, Met can be replaced with Leu or Ile, Phe can be replaced with Met, Leu or Tyr, Ser can be replaced with Thr, Thr can be replaced with Ser, Trp can be replaced with Tyr, Tyr can be replaced with Trp or Phe; and Val can be replaced with Ile or Leu.


Thus, it is understood that, where desired, modifications and changes may be made in the nucleic acid encoding the polypeptides of this invention and/or amino acid sequence of the polypeptides of the present invention and still obtain a polypeptide having like or otherwise desirable characteristics. Such changes may occur in natural isolates or may be synthetically introduced using site-specific mutagenesis, the procedures for which, such as mis-match polymerase chain reaction (PCR), are well known in the art. For example, certain amino acids may be substituted for other amino acids in a polypeptide without appreciable loss of functional activity. It is thus contemplated that various changes may be made in the amino acid sequence of the polypeptides of the present invention (or underlying nucleic acid sequence) without appreciable loss of biological utility or activity and possibly with an increase in such utility or activity. Thus, it is clear that naturally occurring variations in the polypeptide sequences set forth herein as well as genetically engineered variations in the polypeptide sequences set forth herein are contemplated by the present invention. Cells expressing variant polypeptides, whether naturally occurring or genetically engineered can be utilized in the methods of the present invention. By providing the genomic location of genes that are involved in viral infection, the present invention has also provided the genomic location of any variant sequences of these genes. Thus, based on the information provided herein, it would be routine for one of skill in the art to identify and sequence the genomic region identified by applicants and identify variant sequences of the genes set forth herein. It would also be routine for one of skill in the art to utilize comparison tools and bioinformatics techniques to identify sequences from other species that are homologs of the genes set forth herein and are also necessary for infection, but not necessary for survival of the cell.


Methods of Decreasing Infection

The present invention provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 1.


Also provided by the present invention is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 1.


As stated above, an infection can be a bacterial infection, viral infection, fungal infection or a parasitic infection, to name a few. A decrease or inhibition of infection can occur in a cell, in vitro, ex vivo or in vivo. As utilized throughout, the term “infection” encompasses all phases of pathogenic life cycles including, but not limited to, attachment to cellular receptors, entry, internalization, disassembly, replication, genomic integration of pathogenic sequences, transcription of viral RNA, translation of viral RNA, transcription of host cell mRNA, translation of host cell mRNA, proteolytic cleavage of pathogenic proteins or cellular proteins, assembly of particles, endocytosis, cell lysis, budding, and egress of the pathogen from the cells. Therefore, a decrease in infection can be a decrease in attachment to cellular receptors, a decrease in entry, a decrease in internalization, a decrease in disassembly, a decrease in replication, a decrease in genomic integration of pathogenic sequences, decrease in transcription of viral RNA, a decrease in translation of viral RNA, a decrease in transcription of host cell mRNA, a decrease in translation of host cell mRNA, a decrease in proteolytic cleavage of pathogenic proteins or cellular proteins, a decrease in assembly of particles, a decrease in endocytosis, a decrease in cell lysis, a decrease in budding, or a decrease in egress of the pathogen from the cells. This decrease does not have to be complete as this can range from a slight decrease to complete ablation of the infection. A decrease in infection can be at least about 10%, 20%, 30%, 40%, 50%, 60, 70%, 80%, 90%, 95%, 100% or any other percentage decrease in between these percentages as compared to the level of infection in a control cell, for example, a cell wherein expression or activity of Table 1 has not been decreased. A decrease in infection can be at least about 10%, 20%, 30%, 40%, 50%, 60, 70%, 80%, 90%, 95%, 100% or any other percentage decrease in between these percentages as compared to the level of infection in a control cell that has not been contacted with a compound that decreases expression or activity of a gene or gene product set forth in Table 1.


In the methods set forth herein, expression of any gene product of the genes of Table 1 can be inhibited, for example, by inhibiting transcription of the gene, or inhibiting translation of its gene product. Similarly, the activity of a gene product (for example, an mRNA, a polypeptide or a protein) can be inhibited, either directly or indirectly. Inhibition or a decrease in expression does not have to be complete as this can range from a slight decrease in expression to complete ablation of expression. For example, expression can be inhibited by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein the expression of the gene or gene product of Table 1 has not been decreased or inhibited.


In the methods set forth herein, expression can be inhibited, for example, by inhibiting transcription of the gene, or inhibiting translation of its gene product. Similarly, the activity of a gene product (for example, an mRNA, a polypeptide or a protein) can be inhibited, either directly or indirectly. Inhibition or a decrease in expression does not have to be complete as this can range from a slight decrease in expression to complete ablation of expression. For example, expression can be inhibited by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein the expression of a gene or gene product set forth in Table 1 has not been decreased or inhibited.


Similarly, inhibition or decrease in the activity of a gene product does not have to be complete as this can range from a slight decrease to complete ablation of the activity of the gene product. For example, the activity of a gene product can be inhibited by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein activity of a gene or gene product set forth in Table 1 has not been decreased or inhibited. As utilized herein, “activity of a gene product” can be an activity that is involved in pathogenicity, for example, interacting directly or indirectly, with pathogen, e.g. viral protein or viral nucleic acids, or an activity that the gene product performs in a normal cell, i.e. in a non-infected cell. Depending on the gene product, one of skill in the art would know how to assay for an activity that is involved in pathogenicity, an activity that is involved in normal cellular function, or both. As set forth above, an activity of the proteins and nucleic acids listed herein can be the ability to bind or interact with other proteins. Therefore, the present invention also provides a method of decreasing infection by inhibiting or decreasing the interaction between any of the proteins of the present invention and other cellular proteins, such as, for example, receptors, enzymes, nucleic acids and hormones, provided that such inhibition correlates with decreasing infection by the pathogen. Also provided is a method of decreasing infection by inhibiting or decreasing the interaction between any of the proteins of the present invention and a viral, bacterial, parasitic or fungal protein (i.e. a non-host protein).


The cells of the present invention can be prokaryotic or eukaryotic, such as a cell from an insect, fish, crustacean, mammal, bird, reptile, yeast or a bacterium, such as E. coli. The cell can be part of an organism, or part of a cell culture, such as a culture of mammalian cells or a bacterial culture. Therefore, the cell can also be part of a population of cells. The cell(s) can also be in a subject.


Examples of viral infections include but are not limited to, infections caused by RNA viruses (including negative stranded RNA viruses, positive stranded RNA viruses, double stranded RNA viruses and retroviruses) and DNA viruses. All strains, types, subtypes of DNA and RNA viruses are contemplated herein.


Examples of RNA viruses include, but are not limited to picornaviruses, which include aphthoviruses (for example, foot and mouth disease virus 0, A, C, Asia 1, SAT1, SAT2 and SAT3), cardioviruses (for example, encephalomycarditis virus and Theiller's murine encephalomyelitis virus), enteroviruses (for example polioviruses 1, 2 and 3, human enteroviruses A-D, bovine enteroviruses 1 and 2, human coxsackieviruses A1-A22 and A24, human coxsackieviruses B1-B5, human echoviruses 1-7, 9, 11-12, 24, 27, 29-33, human enteroviruses 68-71, porcine enteroviruses 8-10 and simian enteroviruses 1-18), erboviruses (for example, equine rhinitis virus), hepatovirus (for example human hepatitis A virus and simian hepatitis A virus), kobuviruses (for example, bovine kobuvirus and Aichi virus), parechoviruses (for example, human parechovirus 1 and human parechovirus 2), rhinovirus (for example, rhinovirus A, rhinovirus B, rhinovirus C, HRV16, HRV16 (VR-11757), HRV14 (VR-284), or HRV1A (VR-1559), human rhinovirus 1-100 and bovine rhinoviruses 1-3) and teschoviruses (for example, porcine teschovirus).


Additional examples of RNA viruses include caliciviruses, which include noroviruses (for example, Norwalk virus), sapoviruses (for example, Sapporo virus), lagoviruses (for example, rabbit hemorrhagic disease virus and European brown hare syndrome) and vesiviruses (for example vesicular exanthema of swine virus and feline calicivirus).


Other RNA viruses include astroviruses, which include mamastorviruses and avastroviruses. Togaviruses are also RNA viruses. Togaviruses include alphaviruses (for example, Chikungunya virus, Sindbis virus, Semliki Forest virus, Western equine encephalitis, Getah virus, Everglades virus, Venezuelan equine encephalitis virus and Aura virus) and rubella viruses. Additional examples of RNA viruses include the flaviviruses (for example, tick-borne encephalitis virus, Tyuleniy virus, Aroa virus, Dengue virus (types 1 to 4), Kedougou virus, Japanese encephalitis virus (JEV), West Nile virus (WNV), Kokobera virus, Ntaya virus, Spondweni virus, Yellow fever virus, Entebbe bat virus, Modoc virus, Rio Bravo virus, Cell fusing agent virus, pestivirus, GB virus A, GBV-A like viruses, GB virus C, Hepatitis G virus, hepacivirus (hepatitis C virus (HCV)) all six genotypes), bovine viral diarrhea virus (BVDV) types 1 and 2, and GB virus B).


Other examples of RNA viruses are the coronaviruses, which include, human respiratory coronaviruses such as SARS-CoV, HCoV-229E, HCoV-NL63 and HCoV-OC43. Coronaviruses also include bat SARS-like CoV, turkey coronavirus, chicken coronavirus, feline coronavirus and canine coronavirus. Additional RNA viruses include arteriviruses (for example, equine arterivirus, porcine reproductive and respiratory syndrome virus, lactate dehyrogenase elevating virus of mice and simian hemorraghic fever virus). Other RNA viruses include the rhabdoviruses, which include lyssaviruses (for example, rabies, Lagos bat virus, Mokola virus, Duvenhage virus and European bat lyssavirus), vesiculoviruses (for example, VSV-Indiana, VSV-New Jersey, VSV-Alagoas, Piry virus, Cocal virus, Maraba virus, Isfahan virus and Chandipura virus), and ephemeroviruses (for example, bovine ephemeral fever virus, Adelaide River virus and Berrimah virus). Additional examples of RNA viruses include the filoviruses. These include the Marburg and Ebola viruses (for example, EBOV-Z, EBOV-S, EBOV-IC and EBOV-R.


The paramyxoviruses are also RNA viruses. Examples of these viruses are the rubulaviruses (for example, mumps, parainfluenza virus 5, human parainfluenza virus type 2, Mapuera virus and porcine rubulavirus), avulaviruses (for example, Newcastle disease virus), respoviruses (for example, Sendai virus, human parainfluenza virus type 1 and type 3, bovine parainfluenza virus type 3), henipaviruses (for example, Hendra virus and Nipah virus), morbilloviruses (for example, measles, Cetacean morvilliirus, Canine distemper virus, Peste-des-petits-ruminants virus, Phocine distemper virus and Rinderpest virus), pneumoviruses (for example, human respiratory syncytial virus A2, B1 and S2, bovine respiratory syncytial virus and pneumonia virus of mice), metapneumoviruses (for example, human metapneumovirus and avian metapneumovirus). Additional paramyxoviruses include Fer-de-Lance virus, Tupaia paramyxovirus, Menangle virus, Tioman virus, Beilong virus, J virus, Mossman virus, Salem virus and Nariva virus. Additional RNA viruses include the orthomyxoviruses.


These viruses include influenza viruses and strains (e.g., influenza A, influenza A strain A/Victoria/3/75, influenza A strain A/Puerto Rico/8/34, influenza A H1N1 (including but not limited to A/WS/33, A/NWS/33 and A/California/04/2009 strains) influenza B, influenza B strain Lee, and influenza C viruses) H2N2, H3N2, H5N1, H7N7, H1N2, H9N2, H7N2, H7N3 and H10N7), as well as avian influenza (for example, strains H5N1, H5N1 Duck/MN/1525/81, H5N2, H7N1, H7N7 and H9N2) thogotoviruses and isaviruses. Orthobunyaviruses (for example, Akabane virus, California encephalitis, Cache Valley virus, Snowshoe hare virus,) nairoviruses (for example, Nairobi sheep virus, Crimean-Congo hemorrhagic fever virus Group and Hughes virus), phleboviruses (for example, Candiru, Punta Toro, Rift Valley Fever, Sandfly Fever, Naples, Toscana, Sicilian and Chagres), and hantaviruses (for example, Hantaan, Dobrava, Seoul, Puumala, Sin Nombre, Bayou, Black Creek Canal, Andes and Thottapalayam) are also RNA viruses. Arenaviruses such as lymphocytic choriomeningitis virus, Lujo virus, Lassa fever virus, Argentine hemorrhagic fever virus, Bolivian hemorrhagic fever virus, Venezuelan hemorrhagic fever virus, SABV and WWAV are also RNA viruses. Borna disease virus is also an RNA virus. Hepatitis D (Delta) virus and hepatitis E are also RNA viruses.


Additional RNA viruses include reoviruses, rotaviruses, birnaviruses, chrysoviruses, cystoviruses, hypoviruses partitiviruses and totoviruses. Orbiviruses such as African horse sickness virus, Blue tongue virus, Changuinola virus, Chenuda virus, Chobar Gorge Corriparta virus, epizootic hemorraghic disease virus, equine encephalosis virus, Eubenangee virus, Ieri virus, Great Island virus, Lebombo virus, Orungo virus, Palyam virus, Peruvian Horse Sickness virus, St. Croix River virus, Umatilla virus, Wad Medani virus, Wallal virus, Warrego virus and Wongorr virus are also RNA viruses.


Retroviruses include alpharetroviruses (for example, Rous sarcoma virus and avian leukemia virus), betaretroviruses (for example, mouse mammary tumor virus, Mason-Pfizer monkey virus and Jaagsiekte sheep retrovirus), gammaretroviruses (for example, murine leukemia virus and feline leukemia virus, deltraretroviruses (for example, human T cell leukemia viruses (HTLV-1, HTLV-2), bovine leukemia virus, STLV-1 and STLV-2), epsilonretriviruses (for example, Walleye dermal sarcoma virus and Walleye epidermal hyperplasia virus 1), reticuloendotheliosis virus (for example, chicken syncytial virus, lentiviruses (for example, human immunodeficiency virus (HIV) type 1, human immunodeficiency virus (HIV) type 2, human immunodeficiency virus (HIV) type 3, simian immunodeficiency virus, equine infectious anemia virus, feline immunodeficiency virus, caprine arthritis encephalitis virus and Visna maedi virus) and spumaviruses (for example, human foamy virus and feline syncytia-forming virus).


Examples of DNA viruses include polyomaviruses (for example, simian virus 40, simian agent 12, BK virus, JC virus, Merkel Cell polyoma virus, bovine polyoma virus and lymphotrophic papovavirus), papillomaviruses (for example, human papillomavirus, bovine papillomavirus, adenoviruses (for example, adenoviruses A-F, canine adenovirus type I, canined adeovirus type 2), circoviruses (for example, porcine circovirus and beak and feather disease virus (BFDV)), parvoviruses (for example, canine parvovirus), erythroviruses (for example, adeno-associated virus types 1-8), betaparvoviruses, amdoviruses, densoviruses, iteraviruses, brevidensoviruses, pefudensoviruses, herpes viruses 1, 2, 3, 4, 5, 6, 7 and 8 (for example, herpes simplex virus 1, herpes simplex virus 2, varicella-zoster virus, Epstein-Barr virus, cytomegalovirus, Kaposi's sarcoma associated herpes virus, human herpes virus-6 variant A, human herpes virus-6 variant B and cercophithecine herpes virus 1 (B virus)), poxviruses (for example, smallpox (variola), cowpox, monkeypox, vaccinia, Uasin Gishu, camelpox, psuedocowpox, pigeonpox, horsepox, fowlpox, turkeypox and swinepox), and hepadnaviruses (for example, hepatitis B and hepatitis B-like viruses). Chimeric viruses comprising portions of more than one viral genome are also contemplated herein.


For animals, in addition to the animal viruses listed above, viruses include, but are not limited to, the animal counterpart to any above listed human virus. The provided compounds can also decrease infection by newly discovered or emerging viruses. Such viruses are continuously updated on http://en.wikipedia.org/wiki/Virus and www.virology.net.


Examples of bacterial infections include, but are not limited to infections caused by the following bacteria: Listeria (sp.), Franscicella tularensis, Mycobacterium tuberculosis, Rickettsia (all types), Ehrlichia, Chlamydia. Further examples of bacteria that can be targeted by the present methods include M. tuberculosis, M. bovis, M. bovis strain BCG, BCG substrains, M. avium, M. intracellulare, M. africanum, M. kansasii, M. marinum, M. ulcerans, M. avium subspecies paratuberculosis, Nocardia asteroides, other Nocardia species, Legionella pneumophila, other Legionella species, Salmonella typhi, other Salmonella species, Shigella species, Yersinia pestis, Pasteurella haemolytica, Pasteurella multocida, other Pasteurella species, Actinobacillus pleuropneumoniae, Listeria monocytogenes, Listeria ivanovii, Brucella abortus, other Brucella species, Cowdria ruminantium, Chlamydia pneumoniae, Chlamydia trachomatis, Chlamydia psittaci, Coxiella burnetti, other Rickettsial species, Ehrlichia species, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes, Streptococcus agalactiae, Bacillus anthracis, Escherichia coli, Vibrio cholerae, Kingella kingae, Campylobacter species, Neiserria meningitidis, Neiserria gonorrhea, Pseudomonas aeruginosa, other Pseudomonas species, Haemophilus influenzae, Haemophilus ducreyi, other Hemophilus species, Clostridium tetani, other Clostridium species, Yersinia enterolitica, and other Yersinia species.


Examples of parasitic infections include, but are not limited to infections caused by the following parasites: Cryptosporidium, Plasmodium (all species), American trypanosomes (T. cruzi), African trypanosomes, Acanthamoeba, Entaoeba histolytica, Angiostrongylus, Anisakis, Ascaris, Babesia, Balantidium, Baylisascaris, lice, ticks, mites, fleas, Capillaria, Clonorchis, Chilomastix mesnili, Cyclspora, Diphyllobothrium, Dipylidium caninum, Fasciola, Giardia, Gnathostoma, Hetetophyes, Hymenolepsis, Isospora, Loa loa, Microsporidia, Naegleria, Toxocara, Onchocerca, Opisthorchis, Paragonimus, Baylisascaris, Strongyloides, Taenia, Trichomonas and Trichuris.


Furthermore, examples of protozoan and fungal species contemplated within the present methods include, but are not limited to, Plasmodium falciparum, other Plasmodium species, Toxoplasma gondii, Pneumocystis carinii, Trypanosoma cruzi, other trypanosomal species, Leishmania donovani, other Leishmania species, Theileria annulata, other Theileria species, Eimeria tenella, other Eimeria species, Histoplasma capsulatum, Cryptococcus neoformans, Blastomyces dermatitidis, Coccidioides immitis, Paracoccidioides brasiliensis, Penicillium marneffei, and Candida species. The provided compounds can also decrease infection by newly discovered or emerging bacteria, parasites or fungi, including multidrug resistant strains of same.


Furthermore, a decrease of expression or activity of a gene provided herein can result in a decrease in infection for two or more pathogens selected from the group consisting of the viruses, bacteria, pathogen and fungi described herein For example, and not to be limiting, this includes two or more viruses, two or more bacteria, two or more parasites, two or more fungi, or combinations thereof.


Further provided by the present invention is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 1, wherein the pathogen is a respiratory virus. Respiratory viruses include, but are not limited to, picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses and adenoviruses. More specifically, and not to be limiting, the respiratory virus can be an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus or a respiratory syncytial virus (RSV) or any strain thereof.


Also provided by the present invention is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 1, wherein the pathogen is a gastrointestinal virus. Gastrointestinal viruses include, but are not limited to, picornaviruses, filoviruses, flaviviruses, calciviruses and reoviruses. More specifically, and not to be limiting, the gastrointestinal virus can be a reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus.


The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 1, wherein the pathogen is a hemorraghic fever virus. These include, but are not limited to, flaviviruses, bunyaviruses, arenaviruses, filoviruses and hantaviruses. More specifically and not to be limiting, the hemorraghic fever virus can be an Ebola virus, a Marburg virus, a Dengue fever virus (types 1-4), a yellow fever virus, a Sin Nombre virus, a Junin virus, a Machupo virus, a Lassa virus, a Rift Valley fever virus, or a Kyasanur forest disease virus.


The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 1, wherein the pathogen is a pox virus, a herpes virus, BVDV, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 1, wherein the pathogen is a pox virus, lymphocytic choriomeningitis virus (LCM), Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus.


The present invention also provides a method of decreasing the toxicity of a toxin in a cell comprising decreasing expression or activity of a gene or gene product set forth in Table 1. The cell can be in vitro, ex vivo or in vivo. Toxins can include, but are not limited to, a bacterial toxin, neurotoxins, such as botulinum neurotoxins, mycotoxins, ricin, Clostridium perfringens toxins, Clostridium difficile toxins, saxitoxins, tetrodotoxins, abrin, conotoxins, Staphlococcal toxins, E. coli toxins, streptococcal toxins, shigatoxins, T-2 toxins, anthrax toxins, chimeric forms of the toxins listed herein, and the like. The decrease in toxicity can be at least about 10%, 20%, 30%, 40%, 50%, 60, 70%, 80%, 90%, 95%, 100% or any other percentage decrease in between these percentages as compared to the level of toxicity in a cell wherein expression or activity of a gene or gene product set forth in Table 1 has not been decreased.


Toxicity can be measured, for example, via a cell viability, apopotosis assay, LDH release assay or cytotoxicity assay (See, for example, Kehl-Fie and St. Geme “Identification and characterization of an RTX toxin in the emerging pathogen Kingella kingae,” J. Bacteriol. 189(2):430-6 (2006) and Kirby “Anthrax Lethal Toxin Induces Human Endothelial cell Apoptosis,” Infection and Immunity 72: 430-439 (2004), both of which are incorporated herein in their entireties by this reference.)


In the methods of the present invention, expression and/or activity of a gene or gene product set forth in Table 1 can be decreased by contacting the cell with any composition that can decrease expression or activity. For example, the composition can comprise a chemical, a small or large molecule (organic or inorganic), a drug, a protein, a peptide, a cDNA, an antibody, a morpholino, a triple helix molecule, an aptamer, an siRNA, a shRNA, an miRNA, an antisense RNA, a ribozyme or any other compound now known or identified in the future that decreases the expression and/or activity of a gene or gene product set forth in Table 1. A decrease in expression or activity can occur by decreasing transcription of mRNA or decreasing translation of RNA. A composition can also be a mixture or “cocktail” of two or more of the compositions described herein.


A decrease in expression and/or activity can also occur by inhibiting the interaction between any of the proteins set forth in Table 1 and other cellular proteins, such as, for example, transcription factors, receptors, nuclear proteins, transporters, microtubules, membrane proteins, enzymes (for example, ATPases, phosphorylases, oxidoreductases, kinases, phosphatases, synthases, lyases, aromatases, helicases, hydrolases, proteases, transferases, nucleases, ligases, reductases and polymerases) and hormones. A decrease in expression and/or activity can also occur by inhibiting or decreasing the interaction between any of the proteins of the present invention and a cellular nucleic acid or a viral nucleic acid. A decrease can also occur by inhibiting or decreasing the interaction, either direct or indirect, between any of the proteins of the present invention and a viral, bacterial, parasitic or fungal protein (i.e. a non-host protein).


A composition can also be single composition or a mixture, cocktail or combination of two or more compositions, for example, two or more compositions selected from the group consisting of chemical, a compound, a small molecule, an inorganic molecule, an organic molecule, an aptamer, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an antisense nucleic acid, and LNA or a ribozyme. The two or more compositions can be the same or different types of compositions. The two or more compositions can decrease expression or activity of the same target or different targets, as one or more genes or gene products set forth in Table 1 can be modulated to effect a decrease in infection. It is understood that two or more compositions comprises three or more, four or more, five or more etc. For example, and not to be limiting two or more compositions can be an two or more compositions comprising an antisense and a small molecule; or two or more antisense molecules; or two or more small molecules; or two or more compositions comprising an siRNA and a small molecule, etc. It is understood that any combination of the types of compositions set forth herein can be utilized in the methods set forth herein.


These compositions can be used alone or in combination with other therapeutic agents such as antiviral compounds, antibacterial agents, antifungal agents, antiparasitic agents, anti-inflammatory agents, anti-cancer agents, etc. All of the compounds described herein can be contacted with a cell in vitro, ex vivo or in vivo.


Examples of antiviral compounds include, but are not limited to, amantadine, rimantadine, ribavirin, zanamavir (Relenza®) and oseltamavir (Tamiflu®) for the treatment of flu and its associated symptoms. Antiviral compounds useful in the treatment of HIV include Combivir® (lamivudine-zidovudine), maraviroc, Crixivan® (indinavir), Emtriva® (emtricitabine), Epivir® (lamivudine), Fortovase® (saquinavir-sg), Hivid® (zalcitabine), Invirase® (saquinavir-hg), Kaletra® (lopinavir-ritonavir), Lexiva™ (fosamprenavir), Norvir® (ritonavir), Retrovir® (zidovudine), Sustiva® (efavirenz), Videx EC®, (didanosine), Videx® (didanosine), Viracept® (nelfinavir), Viramune® (nevirapine), Zerit® (stavudine), Ziagen® (abacavir), Fuzeon® (enfuvirtide), Rescriptor® (delavirdine), Reyataz® (atazanavir), Trizivir® (abacavir-lamivudine-zidovudine), Viread® (tenofovir disoproxil fumarate), Truvada® (tenofovir-emtricitabine), Atripla® (tenofovir-emtricitabine-efavirenz) and Agenerase® (amprenavir). Other antiviral compounds useful in the treatment of Ebola and other filoviruses include ribavirin and cyanovirin-N (CV-N). For the treatment of herpes virus, Zovirax® (acyclovir) is available. Antibacterial agents include, but are not limited to, antibiotics (for example, penicillin and ampicillin), sulfa Drugs and folic acid Analogs, Beta-Lactams, aminoglycosides, tetracyclines, macrolides, lincosamides, streptogramins, fluoroquinolones, rifampin, mupirocin, cycloserine, aminocyclitol and oxazolidinones.


Antifungal agents include, but are not limited to, amphotericin, nystatin, terbinafine, itraconazole, fluconazole, ketoconazole, and griselfulvin.


Antiparasitic agents include, but are not limited to, antihelmintics, antinematodal agents, antiplatyhelmintic agents, antiprotozoal agents, amebicides, antimalarials, antitrichomonal agents, aoccidiostats and trypanocidal agents.


Antibodies

The present invention also provides antibodies that specifically bind to the gene products, proteins and fragments thereof set forth in Table 1. The antibody of the present invention can be a polyclonal antibody or a monoclonal antibody. The antibody of the invention selectively binds a polypeptide. By “selectively binds” or “specifically binds” is meant an antibody binding reaction which is determinative of the presence of the antigen (in the present case, a polypeptide set forth in Table 1 or antigenic fragment thereof among a heterogeneous population of proteins and other biologics). Thus, under designated immunoassay conditions, the specified antibodies bind preferentially to a particular peptide and do not bind in a significant amount to other proteins in the sample. Preferably, selective binding includes binding at about or above 1.5 times assay background and the absence of significant binding is less than 1.5 times assay background.


This invention also contemplates antibodies that compete for binding to natural interactors or ligands to the proteins set forth in Table 1. In other words, the present invention provides antibodies that disrupt interactions between the proteins set forth in Table 1 and their binding partners. For example, an antibody of the present invention can compete with a protein for a binding site (e.g. a receptor) on a cell or the antibody can compete with a protein for binding to another protein or biological molecule, such as a nucleic acid that is under the transcriptional control of a transcription factor set forth in Table 1. An antibody can also disrupt the interaction between a protein set forth in Table 1 and a pathogen, or the product of a pathogen. For example, an antibody can disrupt the interaction between a protein set forth in Table 1 and a viral protein, a bacterial protein, a parasitic protein, a fungal protein or a toxin. The antibody optionally can have either an antagonistic or agonistic function as compared to the antigen. Antibodies that antagonize pathogenic infection are utilized to decrease infection.


Preferably, the antibody binds a polypeptide in vitro, ex vivo or in vivo. Optionally, the antibody of the invention is labeled with a detectable moiety. For example, the detectable moiety can be selected from the group consisting of a fluorescent moiety, an enzyme-linked moiety, a biotin moiety and a radiolabeled moiety. The antibody can be used in techniques or procedures such as diagnostics, screening, or imaging. Anti-idiotypic antibodies and affinity matured antibodies are also considered to be part of the invention.


As used herein, the term “antibody” encompasses chimeric antibodies and hybrid antibodies, with dual or multiple antigen or epitope specificities, and fragments, such as F(ab′)2, Fab′, Fab and the like, including hybrid fragments. Thus, fragments of the antibodies that retain the ability to bind their specific antigens are provided. Such antibodies and fragments can be made by techniques known in the art and can be screened for specificity and activity according to the methods set forth in the Examples and in general methods for producing antibodies and screening antibodies for specificity and activity (See Harlow and Lane. Antibodies, A Laboratory Manual. Cold Spring Harbor Publications, New York, (1988)).


Also included within the meaning of “antibody” are conjugates of antibody fragments and antigen binding proteins (single chain antibodies) as described, for example, in U.S. Pat. No. 4,704,692, the contents of which are hereby incorporated by reference.


Optionally, the antibodies are generated in other species and “humanized” for administration in humans. In one embodiment of the invention, the “humanized” antibody is a human version of the antibody produced by a germ line mutant animal. Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′)2, or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In one embodiment, the present invention provides a humanized version of an antibody, comprising at least one, two, three, four, or up to all CDRs of a monoclonal antibody that specifically binds to a protein or fragment thereof set forth in Table 1. In some instances, corresponding non-human residues replace Fv framework residues of the human immunoglobulin. Humanized antibodies may also comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of or at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)).


Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source that is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such “humanized” antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.


Peptides that inhibit expression or activity of the genes or gene products set forth in Table 1 are also provided herein. Peptide libraries can be screened utilizing the screening methods set forth herein to identify peptides that inhibit activity of any of the genes or gene products set forth in Table 1. These peptides can be derived from a protein that binds to any of the genes or gene products set forth in Table 1. These peptides can be any peptide in a purified or non-purified form, such as peptides made of D- and/or L-configuration amino acids (in, for example, the form of random peptide libraries; see Lam et al., Nature 354:82-4, 1991), phosphopeptides (such as in the form of random or partially degenerate, directed phosphopeptide libraries; see, for example, Songyang et al., Cell 72:767-78, 1993).


Peptides

Peptides that inhibit expression or activity of a gene or a gene product set forth in Table 1 are also provided herein. Peptide libraries can be screened utilizing the screening methods set forth herein to identify peptides that inhibit activity of any of the genes or gene products set forth in Table 1. These peptides can be derived from a protein that binds to any of the genes or gene products set forth in Table 1. These peptides can be any peptide in a purified or non-purified form, such as peptides made of D- and/or L-configuration amino acids (in, for example, the form of random peptide libraries; see Lam et al., Nature 354:82-4, 1991), phosphopeptides (such as in the form of random or partially degenerate, directed phosphopeptide libraries; see, for example, Songyang et al., Cell 72:767-78, 1993).


siRNAs


Short interfering RNAs (siRNAs), also known as small interfering RNAs, are double-stranded RNAs that can induce sequence-specific post-transcriptional gene silencing, thereby decreasing gene expression (See, for example, U.S. Pat. Nos. 6,506,559, 7,056,704, 7,078,196, 6,107,094, 5,898,221, 6,573,099, and European Patent No. 1.144,623, all of which are hereby incorporated in their entireties by this reference). siRNas can be of various lengths as long as they maintain their function. In some examples, siRNA molecules are about 19-23 nucleotides in length, such as at least 21 nucleotides, for example at least 23 nucleotides. In one example, siRNA triggers the specific degradation of homologous RNA molecules, such as mRNAs, within the region of sequence identity between both the siRNA and the target RNA. For example, WO 02/44321 discloses siRNAs capable of sequence-specific degradation of target mRNAs when base-paired with 3′ overhanging ends. The direction of dsRNA processing determines whether a sense or an antisense target RNA can be cleaved by the produced siRNA endonuclease complex. Thus, siRNAs can be used to modulate transcription or translation, for example, by decreasing expression of a gene set forth in Table 1, 2, 3 or 4. The effects of siRNAs have been demonstrated in cells from a variety of organisms, including Drosophila, C. elegans, insects, frogs, plants, fungi, mice and humans (for example, WO 02/44321; Gitlin et al., Nature 418:430-4, 2002; Caplen et al., Proc. Natl. Acad. Sci. 98:9742-9747, 2001; and Elbashir et al., Nature 411:494-8, 2001).


Utilizing sequence analysis tools, one of skill in the art can design siRNAs to specifically target one or more of the genes set forth in Table 1 for decreased gene expression. siRNAs that inhibit or silence gene expression can be obtained from numerous commercial entities that synthesize siRNAs, for example, Ambion Inc. (2130 Woodward Austin, Tex. 78744-1832, USA), Qiagen Inc. (27220 Turnberry Lane, Valencia, Calif. USA) and Dharmacon Inc. (650 Crescent Drive, #100 Lafayette, Colo. 80026, USA). The siRNAs synthesized by Ambion Inc., Qiagen Inc. or Dharmacon Inc, can be readily obtained from these and other entities by providing a GenBank Accession No. for the mRNA of any gene set forth in Table 1, 2, 3 or 4. In addition, siRNAs can be generated by utilizing Invitrogen's BLOCK-ITTM RNAi Designer https://rnaidesigner.invitrogen.com/rnaiexpress. siRNA sequences can comprise a 3′TT overhang and/or additional sequences that allow efficient cloning and expression of the siRNA sequences. siRNA sequences can be cloned into vectors and utilized in vitro, ex vivo or in vivo to decrease gene expression. One of skill in the art would know that it is routine to utilize publicly available algorithms for the design of siRNA to target mRNA sequences. These sequences can then be assayed for inhibition of gene expression in vitro, ex vivo or in vivo.


Examples of siRNA sequences that can be utilized in the methods described herein include, but are not limited, to those set forth below. Specifically, the sense siRNA sequences set forth below and sequences complementary to these sequences can be used alone or in combination with other sequences to inhibit gene expression. Also contemplated are siRNA sequences that are shorter or longer than the sequences set forth below. For example, an siRNA sequence comprising any of the sequences set forth below can be readily generated by adding nucleotides, on one or both ends of the siRNA, that flank these sequences in the full-length mRNA for the gene of interest. Nucleotides can also be removed, from one or both ends of the siRNA to generate shorter siRNA sequences that retain their function. These sequences can comprise a 3′TT overhang and/or additional sequences that allow efficient cloning and expression of the siRNA sequences. All of the sequences disclosed herein can be cloned into vectors and utilized in vitro, ex vivo or in vivo to decrease gene expression. These siRNA sequences are merely exemplary as one of skill in the art would know that it is routine to utilize publicly available algorithms for the design of siRNA to target mRNA sequences. It is understood that any siRNA sequence set forth in the present application also includes disclosure of its reverse complement to produce siRNA duplexes. For example, the disclosure of CACCGCCGUGAAGAUUGGAAUAAUU (SEQ ID NO: 564) also includes the disclosure of AAUUAUUCCAAUCUUCACGGCGGUG (SEQ ID NO: 2608); the disclosure of GAACAGGCCUGGAUGAUCCAGAAAU (SEQ ID NO: 565) also includes the disclosure of AUUUCUGGAUCAUCCAGGCCUGUUC (SEQ ID NO: 2609), etc. These sequences can then be assayed for inhibition of gene expression in vitro, ex vivo or in vivo.











MTAP



(SEQ ID NO: 564)



CACCGCCGUGAAGAUUGGAAUAAUU







(SEQ ID NO: 565)



GAACAGGCCUGGAUGAUCCAGAAAU







(SEQ ID NO: 566)



GGCAAGCCAUCUGAUGCCUUAAUUU







(SEQ ID NO: 567)



CAGCCCGGCGAUAUUGUCAUUAUUG







(SEQ ID NO: 568)



CCGGCGAUAUUGUCAUUAUUGAUCA







AHR



(SEQ ID NO: 569)



CAGCAAAUUUCAGAGAAGGCCUGAA







(SEQ ID NO: 570)



GAGAAUUCUUAUUACAGGCUCUGAA







(SEQ ID NO: 571)



CAGCGUCAGCUACACUGGGCAUUAA







(SEQ ID NO: 572)



CCUUUAAUGGAGAGGUGCUUCAUAU







(SEQ ID NO: 573)



UACUCCACUUCAGCCACCAUCCAUA







AK5



(SEQ ID NO: 574)



GAAGAUCCAGUAGAAUACUUGGAAA







(SEQ ID NO: 575)



CGGAGAUCCUUUCUAAGAAAUGUAA







(SEQ ID NO: 576)



CCAAUCCAUCAAUUCUCCAUAGAAA







(SEQ ID NO: 577)



CAGAACGAUAUGGAUUCCAAUACAU







(SEQ ID NO: 578)



GCAGCAAUAGGAAAUGGAGUCUUAU







AMOTL2



(SEQ ID NO: 579)



CGUUGAGUGAACGGCUCCUUCAGUU







(SEQ ID NO: 580)



CCCACCUCAGUACCCUCAUGUUGUA







(SEQ ID NO: 581)



ACUAGCUCAUGAGACCACCACUGCU







(SEQ ID NO: 582)



CCGCAUUGAGAAGCUGGAAAGCGAA







(SEQ ID NO: 583)



UGCAAGACUUCAACCGGGAUCUUAG







ANKMY2



(SEQ ID NO: 584)



UACUGUCCAAGAAGCUGGAACAUUA







(SEQ ID NO: 585)



UCCUCUAAUGCAUGCAGCAUAUAAA







(SEQ ID NO: 586)



GAGCCGAUGUAAAUUGUCAUCAGCA







(SEQ ID NO: 587)



CCGAUGUAAAUUGUCAUCAGCAUGA







(SEQ ID NO: 588)



CACAGCCCUCAUGUUUGCUGCACUU







ANXA4



(SEQ ID NO: 589)



CGGCACCGAUGAAGACGCCAUUAUU







(SEQ ID NO: 590)



GGCACCGAUGAAGACGCCAUUAUUA







(SEQ ID NO: 591)



ACGCCAUUAUUAGCGUCCUUGCCUA







(SEQ ID NO: 592)



GAGAUCAGGACAGCCUACAAGAGCA







(SEQ ID NO: 593)



CAUCGUUCAUGUUCCAGCGAGUGCU







ARL6IP5



(SEQ ID NO: 594)



GGGUUUCUGAGUCCCUUCAACAUGA







(SEQ ID NO: 595)



CAGCCCACAAUAAAGACGUCCUUCG







(SEQ ID NO: 596)



CCACGACGUUCGUUAUGGUGGUCAU







(SEQ ID NO: 597)



CGACGUUCGUUAUGGUGGUCAUGUU







(SEQ ID NO: 598)



UAUGGUGGUCAUGUUGGCGAGCUAU







ARSA



(SEQ ID NO: 599)



CACAGACUUCUACGUGCCUGUGUCU







(SEQ ID NO: 600)



CAUCGAUUUCUAGGCAUCCCGUACU







(SEQ ID NO: 601)



GACAAUGGACCUGAGACCAUGCGUA







(SEQ ID NO: 602)



UGCCCAAUGUCACCUUGGAUGGCUU







(SEQ ID NO: 603)



CCAAUGUCACCUUGGAUGGCUUUGA







ATOH8



(SEQ ID NO: 604)



UGAAGGAGCUGAACGGCCUUAAGAA







(SEQ ID NO: 605)



ACGGCCUUAAGAAGCUCAAGCGGAA







(SEQ ID NO: 606)



CGGCCUUAAGAAGCUCAAGCGGAAA







(SEQ ID NO: 607)



GGGAAAGUUCCUACUCGUCAAUUUC







(SEQ ID NO: 608)



GGAAAGUUCCUACUCGUCAAUUUCA







ATP6V1A



(SEQ ID NO: 609)



CACAGCGAAUUGGUUGGAGAGAUUA







(SEQ ID NO: 610)



CAGAGAUAUCAAAUGGGACUUUACA







(SEQ ID NO: 611)



CAAAUGGGACUUUACACCUUGCAAA







(SEQ ID NO: 612)



CAAUAUGCCUGUUGCUGCUAGAGAA







(SEQ ID NO: 613)



GCUGCUAGAGAAGCCUCUAUUUAUA







BPNT1



(SEQ ID NO: 614)



GCAGGAAUGAUAGUCAGACGUGUUA







(SEQ ID NO: 615)



CAGGAAUGAUAGUCAGACGUGUUAU







(SEQ ID NO: 616)



GGGUAUUGUGGAGAAGACCUGUGCA







(SEQ ID NO: 617)



CGAUUGGCACAGAUGAGCAUAUGUU







(SEQ ID NO: 618)



CCCAUCGCAGUACAGUGCUAUUAAA







C11ORF54



(SEQ ID NO: 619)



UCUCUGUAGUUGAUUGCCCUGAUUU







(SEQ ID NO: 620)



AGGAACCCUUUACCUUUCCUGUAAA







(SEQ ID NO: 621)



CAGAAGUUGGAGGUGUGCCUUACUU







(SEQ ID NO: 622)



GAAGUUGGAGGUGUGCCUUACUUAU







(SEQ ID NO: 623)



UGCCUUACUUAUUGCCUCUUGUAAA







C170RF75



(SEQ ID NO: 624)



CAGCCACUACUGUCUUUACAGCUAU







(SEQ ID NO: 625)



UCCAGGUUACCGAGUUGGUUGUUAU







(SEQ ID NO: 626)



CCAGGUUACCGAGUUGGUUGUUAUU







(SEQ ID NO: 627)



GAGCAGCUGGUUUGAGGAUGUUGUA







(SEQ ID NO: 628)



GAGGAUGUUGUAUGCCCAAUCCAAA







C180RF32



(SEQ ID NO: 629)



GCAUUCCUUGUAUCGUCAUUCCAGU







(SEQ ID NO: 630)



UCGUCAUUCCAGUUCUGCUCUGGAU







(SEQ ID NO: 631)



UCGUUAGUCGUAUAUGGCCUAAGAA







(SEQ ID NO: 632)



CGUUAGUCGUAUAUGGCCUAAGAAA







(SEQ ID NO: 633)



AAACUUUAAGGGUGCAGACAUGAAU







C1ORF116



(SEQ ID NO: 634)



AGGGUUGCCUCAGAAUGCAAGAGCU







(SEQ ID NO: 635)



CCGGAAGCUGCCACCUAAUAUUGUU







(SEQ ID NO: 636)



AGCUGCCACCUAAUAUUGUUCUGAA







(SEQ ID NO: 637)



CAAGAUGAGCCUGGACUCCACUUAA







(SEQ ID NO: 638)



UCCAGGCAAUGUUGCAGCUAGCAAA







C6ORF176



(SEQ ID NO: 639)



GAGAGAUCUUCUAUCCACAUGGACA







(SEQ ID NO: 640)



GCAGAUGUUCUAUCCAACAUGGAAA







(SEQ ID NO: 641)



UCCAACAUAGGAACCAUGAACUGUA







(SEQ ID NO: 642)



CACAAAGGAAGAUGUUCUAUCCAAA







(SEQ ID NO: 643)



GGGAGAUCUUCUAUCCAAUAUGUAA







C6ORF62



(SEQ ID NO: 644)



CAGGCCAAAUUUGAGUUUCAUCAUG







(SEQ ID NO: 645)



GGCCAAAUUUGAGUUUCAUCAUGGU







(SEQ ID NO: 646)



CCAAAUUUGAGUUUCAUCAUGGUGA







(SEQ ID NO: 647)



UGAGUUUCAUCAUGGUGACUAUGAA







(SEQ ID NO: 648)



GAGUUUCAUCAUGGUGACUAUGAAA







CBLN2



(SEQ ID NO: 649)



CCAUCUAUUUCGACCAGGUAUUAGU







(SEQ ID NO: 650)



CCACUUUGAUCUUGCUUCCAGUAUA







(SEQ ID NO: 651)



CACUUUGAUCUUGCUUCCAGUAUAU







(SEQ ID NO: 652)



CAGACAAACCAUCCAGGUCAGUUUA







(SEQ ID NO: 653)



CCAUCCAGGUCAGUUUAAUGCAGAA







CCDC57



(SEQ ID NO: 654)



ACUGCUGCUGGAGUUUGAAUCGAAA







(SEQ ID NO: 655)



CCUUGUCCCGGGAGCUUAAGGUUAA







(SEQ ID NO: 656)



CCCGGGAGCUUAAGGUUAAACUACU







(SEQ ID NO: 657)



CCAACUUCGUGAAGAUGCAUCAACU







(SEQ ID NO: 658)



CAGCGCCGCUGUGAUGACAUUGAAA







CDH9



(SEQ ID NO: 659)



CAGGUGGAACCGGAAUCGGAAUUUA







(SEQ ID NO: 660)



GAACCGGAAUCGGAAUUUAUCAUUA







(SEQ ID NO: 661)



GAUCCAGAUGCCAGGAACAAUUUAA







(SEQ ID NO: 662)



CGGCAUACUGAUAUGGACCGUAUUU







(SEQ ID NO: 663)



AGCCACAUCCCUGUCUUCAUCAGAA







CEP152



(SEQ ID NO: 664)



CCAAUGGUCAGAAGCAGGAAUUUAA







(SEQ ID NO: 665)



CAGAACAACUGAAAUGGCUCUGGAA







(SEQ ID NO: 666)



CAAAGAAGUACGAAGAGCAAGUAUU







(SEQ ID NO: 667)



CAGCGUUUGCUGGGUAGCAACUCAA







(SEQ ID NO: 668)



UCAGAGAUGAUAUUCUGCUGCUUAA







CKAP2L



(SEQ ID NO: 669)



CAAACAAAGAGAACUUGCUCGAUAU







(SEQ ID NO: 670)



AAGUAAGCCAAAGACUGACUCUUAU







(SEQ ID NO: 671)



CACAAAGCAGGACUUUCCCAGUAAA







(SEQ ID NO: 672)



CCUAAUUUGACAGUUGGCAGAUUUA







(SEQ ID NO: 673)



GCACAGACUUUGGACUCCAAGUUGA







CLTC



(SEQ ID NO: 674)



UCUAGCCUUGCAGGGUGCCAGAUUA







(SEQ ID NO: 675)



GGGAGCUAUGCAGCUAUAUUCUGUA







(SEQ ID NO: 676)



GAAGAACUCUUUGCCCGGAAAUUUA







(SEQ ID NO: 677)



CCAGCCAGGUCAAACUUCUCCUCUA







(SEQ ID NO: 678)



CAGGUCAAACUUCUCCUCUACUUCA







CLUL1



(SEQ ID NO: 679)



GGUGAAUGCAGGUCUUGCCUGGAAA







(SEQ ID NO: 680)



CAUGAGAAUUUAUACAACCUGCCAA







(SEQ ID NO: 681)



AGAAUUUAUACAACCUGCCAACCUA







(SEQ ID NO: 682)



CCAACCUAGCUGGUCCUCUGUGAAA







(SEQ ID NO: 683)



UCAGCCAGUUGACUGUGGAUGUGAA







CNTN5



(SEQ ID NO: 684)



CAGUCACCAUGUGUCUUUCAGAGUA







(SEQ ID NO: 685)



CAGAUGGCUUCGAAAUGGAACAGAA







(SEQ ID NO: 686)



CCACAUUCACCAGAGAUCAUCUAUA







(SEQ ID NO: 687)



GAGAUCAUCUAUAGCUGGGUAUUUA







(SEQ ID NO: 688)



CAAACAUCAGAUGUUGGCAGCUAUA







CRYZ



(SEQ ID NO: 689)



CCUGAAAUUGCGAUCAGAUAUUGCA







(SEQ ID NO: 690)



CAGAUAUUGCAGUACCGAUUCCAAA







(SEQ ID NO: 691)



GACAUACAUUCGCUCUGGUACUUAU







(SEQ ID NO: 692)



GAUAAUGCAUCUGCUUUCAAGAAAG







(SEQ ID NO: 693)



GCUCUUGCAGCAGACCACACUGUUU







DAB2



(SEQ ID NO: 694)



GCCAAGACUCUAUGAUGAAACUAAA







(SEQ ID NO: 695)



GGUCAGCAAUUUGACCAGAUCUCUA







(SEQ ID NO: 696)



CAGGAAGAGGCAGAAGGACUGCUAA







(SEQ ID NO: 697)



GAGCAGACUUCUUCUGGGACUUUGA







(SEQ ID NO: 698)



CAGACCAUGAUGACUUUGAUGCUAA







DARS2



(SEQ ID NO: 699)



GCCAACAGGUGAGAUUGAAAUCAAA







(SEQ ID NO: 700)



AAUAUCUCUGUAAUCUGCAUGGGUU







(SEQ ID NO: 701)



CCGAUGUUAUCGAGAUGAAGGUUCA







(SEQ ID NO: 702)



CAAGACCAGACAGACAGCCUGAGUU







(SEQ ID NO: 703)



CAGACAGCCUGAGUUUACUCAGAUU







DDIT4



(SEQ ID NO: 704)



GGAUGAACACUUGUGUGCCAACCUG







(SEQ ID NO: 705)



GAUGAACACUUGUGUGCCAACCUGA







(SEQ ID NO: 706)



ACUUGUGUGCCAACCUGAUGCAGCU







(SEQ ID NO: 707)



UGCUGAUGCCUAGCCAGUUGGUAAG







(SEQ ID NO: 708)



UGGUAAGCCAGGUGGGCAAAGAACU







DDX42



(SEQ ID NO: 709)



GACCAGGAAGUAGCUUUGCUCAUUU







(SEQ ID NO: 710)



CAGCCUUCAUUUGGCCCAUGUUGAU







(SEQ ID NO: 711)



CAGGUCGACUGAUAGAUCAUGUGAA







(SEQ ID NO: 712)



CAGAGACAUCCUGAUCGACCCUAUU







(SEQ ID NO: 713)



GCGUCUGGUAGAAUUUACCUCUUCA







DNAH2



(SEQ ID NO: 714)



CAUACACGAGCAAUUUGCCAUUCUU







(SEQ ID NO: 715)



GCGGGAGUUUGAUCAGGAAUCUGAA







(SEQ ID NO: 716)



GCUGCCCAAUGAAUCGACCUUAUUU







(SEQ ID NO: 717)



GAGAUGACAUUUGUGUUCAGCAUGA







(SEQ ID NO: 718)



GCACCAUGAUCAAUCAGAAGCUUCA







DUSP5



(SEQ ID NO: 719)



CCUCUACCUUGGAAGUGCCUACCAU







(SEQ ID NO: 720)



UACCUUGGAAGUGCCUACCAUGCAU







(SEQ ID NO: 721)



GCGACCCACCUACACUACAAAUGGA







(SEQ ID NO: 722)



CAAGAAGCAAUAGACUUCAUUGACU







(SEQ ID NO: 723)



AGACUUCAUUGACUGUGUCAGGGAA







EBNA1BP2



(SEQ ID NO: 724)



CAGACAGAGAGUUGCAGGAUGCGUU







(SEQ ID NO: 725)



CAGCACCUCAGAACAAGGACCAGAA







(SEQ ID NO: 726)



GAACAAGGACCAGAAAGCUGUUGAU







(SEQ ID NO: 727)



CAGAAGACGACUUCCAGCGAGAGAU







(SEQ ID NO: 728)



CCCUACGAAGCGACCCACUGAUUAU







EEF1A1



(SEQ ID NO: 729)



AAGUGAAUCUUUGGAAACAAA







(SEQ ID NO: 730)



CACCUGUAAGAUUUACCAGUA







(SEQ ID NO: 731)



AAGGAAUAUCAUUUAAAGCUA







(SEQ ID NO: 732)



CAAGUCUGUAAUGAAGUGUUA







EID3



(SEQ ID NO: 733)



CCGGUUUCUUGUUAUGGCUUCUGAU







(SEQ ID NO: 734)



CGGUUUCUUGUUAUGGCUUCUGAUU







(SEQ ID NO: 735)



GGGUCUGAAUUGGAUGGAAGGCGAU







(SEQ ID NO: 736)



UCCUGACAAGUUGAGUGAUUGUGAU







(SEQ ID NO: 737)



AAGGAAGCAACAUCCUGGAUGGUAA







ENO1



(SEQ ID NO: 738)



CCCACUGUUGAGGUUGAUCUCUUCA







(SEQ ID NO: 739)



CAGUGGUGCUUCAACUGGUAUCUAU







(SEQ ID NO: 740)



CCGGGACAAUGAUAAGACUCGCUAU







(SEQ ID NO: 741)



CGGGACAAUGAUAAGACUCGCUAUA







(SEQ ID NO: 742)



UCAAAGGCUGUUGAGCACAUCAAUA







ERGIC1



(SEQ ID NO: 743)



CCUCUCGGAGCUCACCGGAUUUAUA







(SEQ ID NO: 744)



CACCGGAUUUAUAACGACAGAAGUU







(SEQ ID NO: 745)



CGACGUCAGUCUGAACAUCAGUUUA







(SEQ ID NO: 746)



GCGAGUUGGUUGGGCUUGACAUUCA







(SEQ ID NO: 747)



CCAGAACAUCCACGGAGCUUUCAAU







FAM126B



(SEQ ID NO: 748)



GAUCACCAGUUAUGCAGCAACUUUA







(SEQ ID NO: 749)



CAUCAGCUGUUUGAGCUCUAUCGUA







(SEQ ID NO: 750)



GCCAGAAUUGAUGUGGGUUUAUUUA







(SEQ ID NO: 751)



CAGACAGAGUAAUGGUUGCAUUGAA







(SEQ ID NO: 752)



CAGAACCGGUUUGAAGUCCUGAGUU







FAM35A



(SEQ ID NO: 753)



CCAGUGUAGUUAGUGAAGUUGUACU







(SEQ ID NO: 754)



GGAGCACCUUCAACCUGAUGUAUUA







(SEQ ID NO: 755)



ACGCAGUACUAAGAGUUGUUGAUUU







(SEQ ID NO: 756)



CCAAGAUCAACAUUAUGCGCUUGUA







(SEQ ID NO: 757)



CAACAUCGUAUAUACUGGUUGUGCA







FAM55C



(SEQ ID NO: 758)



CCGAAAUCCCUACUGUGGCUAUGAU







(SEQ ID NO: 759)



CAUCUUGAACUCUGCUGCCUUCUUU







(SEQ ID NO: 760)



GGGUGGUGGAUUACCAGAAUGGGUU







(SEQ ID NO: 761)



CAACUCCAGUGGACCUGAUUGGGUA







(SEQ ID NO: 762)



GACCAGUGGAGGCCCAGAAAGUUUA







FKBP2



(SEQ ID NO: 763)



CCACUGUCCCAUCAAAUCGCGCAAA







(SEQ ID NO: 764)



AAUAGAGCGACGAACUGAGCUGUAA







FNDC3A



(SEQ ID NO: 765)



UCUACACAUGGAAGGUCCAACUUUA







(SEQ ID NO: 766)



CAGCCUCAUUAAUGGUGAAACAGAU







(SEQ ID NO: 767)



CAGAUGAAAGUAGUGUACCAGAGCU







(SEQ ID NO: 768)



CCACCUUGAGCUGUGAACCUGAUAU







(SEQ ID NO: 769)



GGGAACAUCUUUGUGAUCGACUGAA







FNDC3B



(SEQ ID NO: 770)



UCCCAUUCAUGUGCCUCCAGGUUAU







(SEQ ID NO: 771)



CGAAUGAUUCAGACUUGCAAGAAUA







(SEQ ID NO: 772)



CAGGCAAGAGCAGUUGUGUUGUCCU







(SEQ ID NO: 773)



CAUGUGAGGGUGUAUGCCAUGUACA







(SEQ ID NO: 774)



CCAAGAGGUGGUGUGCUACACAUUA







G2E3



(SEQ ID NO: 775)



UAGAAGAUAUCAGGAAGGAAGUGAA







(SEQ ID NO: 776)



GAAGGAAGUGAAUAGAGCUUCUAAA







(SEQ ID NO: 777)



CAUGCACCAUUUGCUUGGAAUUUAU







(SEQ ID NO: 778)



CGCUUGGUUUCAUAGAGACUGUUUA







(SEQ ID NO: 779)



GAGAUGUUGAGAAUGGGAAUUCAUA







GDF15



(SEQ ID NO: 780)



CAAGAACUCAGGACGGUGAAUGGCU







(SEQ ID NO: 781)



UCAGGACGGUGAAUGGCUCUCAGAU







(SEQ ID NO: 782)



UCGGACCAACUGCUGGCAGAAUCUU







(SEQ ID NO: 783)



GACCAACUGCUGGCAGAAUCUUCGU







GEN1



(SEQ ID NO: 784)



CCAGAUGAAGUAAUGAGCUUUCAGU







(SEQ ID NO: 785)



UCUGCCUCAUUGAAUAGCUUGCUUU







(SEQ ID NO: 786)



CACUAUUGACUGGGAAGGUACUUCU







(SEQ ID NO: 787)



UAUCCCAGAACAACUGUCCUGUGAA







(SEQ ID NO: 788)



GAAAGUGUUGGAUGAGGAUUCUGAU







GLIS2



(SEQ ID NO: 789)



UGGGCCAAGUGUAACCAGCUCUUUG







(SEQ ID NO: 790)



GGGCCAAGUGUAACCAGCUCUUUGA







(SEQ ID NO: 791)



UGCAAGACCUGGUGGACCAUGUCAA







(SEQ ID NO: 792)



GACCUGGUGGACCAUGUCAACGAUU







(SEQ ID NO: 793)



GACCAUGUCAACGAUUACCAUGUCA







GLUD1



(SEQ ID NO: 794)



GCCAGCACCAUAGGGCACUAUGAUA







(SEQ ID NO: 795)



CCAGCACCAUAGGGCACUAUGAUAU







(SEQ ID NO: 796)



CACCAUAGGGCACUAUGAUAUUAAU







(SEQ ID NO: 797)



GCUCAUGUCUGUUCAAGAGAGUUUA







(SEQ ID NO: 798)



CAUGUCUGUUCAAGAGAGUUUAGAA







GLYCTK



(SEQ ID NO: 799)



GACCGGAACUUUCAGCUGAGGCAAA







(SEQ ID NO: 800)



CCAGUUCCCACAAUGUGCAAGAUUG







(SEQ ID NO: 801)



UGCCACGUUCUGUGAAGACUGUGCU







(SEQ ID NO: 802)



CCACGUUCUGUGAAGACUGUGCUGU







(SEQ ID NO: 803)



ACGUUCUGUGAAGACUGUGCUGUCU







GTF2H5



(SEQ ID NO: 804)



GGUCAACGUCUUGAAAGGAGUGCUU







(SEQ ID NO: 805)



UCAACGUCUUGAAAGGAGUGCUUAU







(SEQ ID NO: 806)



CAACGUCUUGAAAGGAGUGCUUAUA







(SEQ ID NO: 807)



CGUCUUGAAAGGAGUGCUUAUAGAA







(SEQ ID NO: 808)



GAAAGGAGUGCUUAUAGAAUGUGAU







HAVCR1



(SEQ ID NO: 809)



CAGUGGCGUAUAUUGUUGCCGUGUU







(SEQ ID NO: 810)



CCGUGGGUGGUUCAAUGACAUGAAA







(SEQ ID NO: 811)



GACAACAACUGUCUCUACCUUUGUU







(SEQ ID NO: 812)



GAACCAUGAACCAGUAGCCACUUCA







(SEQ ID NO: 813)



GAGAACCCACCAGCUCACCAUUGUA







HLA-DMA



(SEQ ID NO: 814)



CCAAUGUGGCCAGAUGACCUGCAAA







(SEQ ID NO: 815)



CGAAUUUGCUGACUGGGCUCAGGAA







(SEQ ID NO: 816)



GCGAGUGGAUGAUCCAGCAAAUAGG







(SEQ ID NO: 817)



GAGGGUUUCCUAUCGCUGAAGUGUU



(SEQ ID NO: 818)







CCUAUCGCUGAAGUGUUCACGCUGA







HNRNPH3



(SEQ ID NO: 819)



CAGUACGACUUCGUGGACUACCAUU







(SEQ ID NO: 820)



GGACUACCAUUUGGUUGCAGCAAAG







(SEQ ID NO: 821)



GGCCUUCGUGCAGUUUGCUUCAAAG







(SEQ ID NO: 822)



CAGAAGUAGCAGGAGUGAAAUCAAA







(SEQ ID NO: 823)



GGGACCAUAUGAUAGACCAAUAGGA







HNRNPK



(SEQ ID NO: 824)



CCGACCGGCUGAGGCGGCGCGGCAG







(SEQ ID NO: 825)



CCGGCUGAGGCGGCGCGGCAGCGGA







(SEQ ID NO: 826)



CGGCUGAGGCGGCGCGGCAGCGGAG







HSP90AB4P



(SEQ ID NO: 827)



CCUUCAGGAGUUGAUCUCUAAUGCU







(SEQ ID NO: 828)



CCCUUCCAAGUUGGACGGUGGUAAA







(SEQ ID NO: 829)



AGCCAUUGCCAAGUCUGGUACUGAA







(SEQ ID NO: 830)



CCAAGUCUGGUACUGAAGCAUUUAU







(SEQ ID NO: 831)



CACCCUCAGUUCAUAGGCUGUCUUA







IARS2



(SEQ ID NO: 832)



CAAAUGUAUGAUAAGGGCUUGGUUU







(SEQ ID NO: 833)



CCUCUACGUACUGGCGGCAGAUAAA







(SEQ ID NO: 834)



GGCGGCAGAUAAAGUAGCAUCUGUU







(SEQ ID NO: 835)



CAGUGGUUUAUAAACAUCACGGAUA







(SEQ ID NO: 836)



GAGCAGAUUUGUACUUGGAAGGAAA







IL18



(SEQ ID NO: 837)



GCUGAACCAGUAGAAGACAAUUGCA







(SEQ ID NO: 838)



CCAGUAGAAGACAAUUGCAUCAACU







(SEQ ID NO: 839)



UGCAUCAACUUUGUGGCAAUGAAAU







(SEQ ID NO: 840)



AAGUUCUCUUCAUUGACCAAGGAAA







(SEQ ID NO: 841)



CCAAGGAAAUCGGCCUCUAUUUGAA







IL6ST



(SEQ ID NO: 842)



UCGGACAGCUUGAACAGAAUGUUUA







(SEQ ID NO: 843)



CACUUGGAGACAAACUUCACUUUAA







(SEQ ID NO: 844)



CACAAGUUUGCUGAUUGCAAAGCAA







(SEQ ID NO: 845)



AAGUGAAGCCCAAUCCGCCACAUAA







(SEQ ID NO: 846)



CCAACCCAAGUAUUAAGAGUGUUAU







ITGB1



(SEQ ID NO: 847)



AAAAGUCUUGGAACAGAUCUG







(SEQ ID NO: 848)



AAGAGGGAUAAUACAAAUGAA







(SEQ ID NO: 849)



ACAGAUGAAGUUAACAGUGAA







(SEQ ID NO: 850)



CUGCGAGUGUGAUAAUUUCAA







KDM6B



(SEQ ID NO: 851)



GGGAAGUUUCGAGAGUCCUACCUUU







(SEQ ID NO: 852)



CACCCAGCAUCUAUCUGGAGAGCAA







(SEQ ID NO: 853)



CAGACCCGAAGAACCAUCACAUCAU







(SEQ ID NO: 854)



CAACAUCGACUUGUCUGAUGCUAAG







(SEQ ID NO: 855)



GGCCACCAGGAGAAUAACAACUUCU







KAZALD1



(SEQ ID NO: 856)



CCCAGAUCGUGUCACAUCCAUAUGA







(SEQ ID NO: 857)



CAGAUCGUGUCACAUCCAUAUGACA







(SEQ ID NO: 858)



UGUCACAUCCAUAUGACACUUGGAA







(SEQ ID NO: 859)



CACUUGGAAUGUGACAGGGCAGGAU







(SEQ ID NO: 860)



GGAAUGUGACAGGGCAGGAUGUGAU







KCTD1



(SEQ ID NO: 861)



CAUCCCUACUCCAGCACAACUCACA







(SEQ ID NO: 862)



UCCCUACUCCAGCACAACUCACAAA







(SEQ ID NO: 863)



UCCAAUGCGCCUGUCCACAUUGAUG







(SEQ ID NO: 864)



CCAAUGCGCCUGUCCACAUUGAUGU







(SEQ ID NO: 865)



CCUCACCAAAUACCCUGAAUCCAGA







KIAA1199



(SEQ ID NO: 866)



ACCGUGGAGUUAUUGUUCAUGUCAU







(SEQ ID NO: 867)



CACAGUCAUCCAUUCUGACCGGUUU







(SEQ ID NO: 868)



GAGAGUGAACGUCUGGUCCAGUAUU







(SEQ ID NO: 869)



GGACGGAGUGGUUCGAUCAUGAUAA







(SEQ ID NO: 870)



ACCACUGUCUUGGCCUCCUUGUCAA







KYNU



(SEQ ID NO: 871)



CAGCGCAUUGCGGCUGAACUCAAAU







(SEQ ID NO: 872)



AAUACAGGAUCUGCCUCCAGUUGAU







(SEQ ID NO: 873)



GAAAUUCUCUUGGCCUUCAACCAAA







(SEQ ID NO: 874)



GAGAUGAGAGUAUUGUAGGCCUUAU







(SEQ ID NO: 875)



CCCUUCUGAUCAUUAUGCUAUUGAG







LMX1B



(SEQ ID NO: 876)



GCUACUUCCGGGAUCGGAAACUGUA







(SEQ ID NO: 877)



CCGGGAUCGGAAACUGUACUGCAAA







(SEQ ID NO: 878)



GAUCGGAAACUGUACUGCAAACAAG







(SEQ ID NO: 879)



GGAAACUGUACUGCAAACAAGACUA







(SEQ ID NO: 880)



GCAAACAAGACUACCAACAGCUCUU







LNX2



(SEQ ID NO: 881)



GAUGAUGACCUAGUCUGCCAUAUUU







(SEQ ID NO: 882)



GCAAGAAGUCUAGUAUUCUAGUUCA







(SEQ ID NO: 883)



CACCACACAACAGCCACUUAGUUUA







(SEQ ID NO: 884)



GGGUGGCAACGAAACACCUUUGAUU







(SEQ ID NO: 885)



CAGAUUCUUCAGGUCAACAACUACA







MALAT1



(SEQ ID NO: 886)



GAGAGAUGAGUUGGGAUCAAGUGGA







(SEQ ID NO: 887)



GAGUUGGGAUCAAGUGGAUUGAGGA







(SEQ ID NO: 888)



CCUCAGACAGGUAUCUCUUCGUUAU







(SEQ ID NO: 889)



CGUUAUCAGAAGAGUUGCUUCAUUU







(SEQ ID NO: 890)



CAGGCAGCUGUUAACAGAUAAGUUU







MAPK1IP1L



(SEQ ID NO: 891)



GGCGCUUCCUGUUCCGGCGCCAGGA







(SEQ ID NO: 892)



GCUUCCUGUUCCGGCGCCAGGAGGA







MAP4



(SEQ ID NO: 893)



CCAGAUUGAAGAUACUCCAUCUUCU







(SEQ ID NO: 894)



CGAUACUACAGGGUCUCCAACUGAA







(SEQ ID NO: 895)



CAGGAAUACCCAAAUAGCCAGAACU







(SEQ ID NO: 896)



CGAUCCUAUCCAGACUGAUCCCUUU







(SEQ ID NO: 897)



UCUCCACACUCAGAGUCCUUUGUUU







MGAT3



(SEQ ID NO: 898)



AGAUGAGACGCUACAAGCUCUUUCU







(SEQ ID NO: 899)



UGAGACGCUACAAGCUCUUUCUCAU







(SEQ ID NO: 900)



CGCUACAAGCUCUUUCUCAUGUUCU







(SEQ ID NO: 901)



UACAAGCUCUUUCUCAUGUUCUGUA







(SEQ ID NO: 902)



CAAGCUCUUUCUCAUGUUCUGUAUG







MICALCL



(SEQ ID NO: 903)



CAGUGUUUCUGGGAAUGCUCCAGAU







(SEQ ID NO: 904)



AGACCCUGCAGAGAUGACUUCUGAU







(SEQ ID NO: 905)



GAAGACCAUGUUGUUGGAUUGGAAU







(SEQ ID NO: 906)



UCGAGAAAGUGAGUUUGCAAGAGAA







(SEQ ID NO: 907)



CAGAUGCUUCUAAGCCUCCAAAGAA







MIR194-1



(SEQ ID NO: 908)



GGUGUUAUCAAGUGUAACAGCAACU







MIR215



(SEQ ID NO: 909)



AAAUGACCUAUGAAUUGACAGACAA







(SEQ ID NO: 910)



AAUGACCUAUGAAUUGACAGACAAU







(SEQ ID NO: 911)



GACCUAUGAAUUGACAGACAAUAUA







(SEQ ID NO: 912)



CAAUAUAGCUGAGUUUGUCUGUCAU







(SEQ ID NO: 913)



UAGCUGAGUUUGUCUGUCAUUUCUU







MIR632



(SEQ ID NO: 914)



CCUCCUACCGCAGUGCUUGACGGGA







(SEQ ID NO: 915)



CCGCAGUGCUUGACGGGAGGCGGAG







MIRLET7G



(SEQ ID NO: 916)



GGCUGAGGUAGUAGUUUGUACAGUU







(SEQ ID NO: 917)



GCUGAGGUAGUAGUUUGUACAGUUU







(SEQ ID NO: 918)



GAGGUAGUAGUUUGUACAGUUUGAG







(SEQ ID NO: 919)



GGUAGUAGUUUGUACAGUUUGAGGG







MIRN15A



(SEQ ID NO: 920)



CCUUGGAGUAAAGUAGCAGCACAUA







(SEQ ID NO: 921)



UGGAGUAAAGUAGCAGCACAUAAUG







(SEQ ID NO: 922)



GAGUAAAGUAGCAGCACAUAAUGGU







(SEQ ID NO: 923)



AGUAAAGUAGCAGCACAUAAUGGUU







MIRN16-1



(SEQ ID NO: 924)



UCAGCAGUGCCUUAGCAGCACGUAA







(SEQ ID NO: 925)



CAGCAGUGCCUUAGCAGCACGUAAA







(SEQ ID NO: 926)



AGCAGUGCCUUAGCAGCACGUAAAU







(SEQ ID NO: 927)



GCAGUGCCUUAGCAGCACGUAAAUA







(SEQ ID NO: 928)



CAGUGCCUUAGCAGCACGUAAAUAU







MIRN16-2



(SEQ ID NO: 929)



UCCACUCUAGCAGCACGUAAAUAUU







(SEQ ID NO: 930)



CCACUCUAGCAGCACGUAAAUAUUG







(SEQ ID NO: 931)



GCAGCACGUAAAUAUUGGCGUAGUG







(SEQ ID NO: 932)



AGCACGUAAAUAUUGGCGUAGUGAA







(SEQ ID NO: 933)



GCACGUAAAUAUUGGCGUAGUGAAA







MLF1IP



(SEQ ID NO: 934)



CGUGUUCGACUUUCCUGAUAAUUCU







(SEQ ID NO: 935)



UAGCACAGCUAUAUAUGCUGAUGAA







(SEQ ID NO: 936)



CACUUCUGGAAAUGAAGCAAGUGAA







(SEQ ID NO: 937)



UGAAGCAAGUGAAAUCGAAUCUGUA







(SEQ ID NO: 938)



CAGGCCCAUUAGUGAUGACUCUGAA







MLL5



(SEQ ID NO: 939)



CAGUUUGAAGCAAAUGGGUAUUUCU







(SEQ ID NO: 940)



AGACACAGAACUGUCAGCAUGUGUU







(SEQ ID NO: 941)



CCACUACUAUUAAAUGACAGCUGUU







(SEQ ID NO: 942)



UAUCUGGUUUCGGACGGACUGUUAA







(SEQ ID NO: 943)



GGACGGACUGUUAAUGACAAUUUGA







MRPL45



(SEQ ID NO: 944)



CAUGACCGACUUCAUACCUUGGUAA







(SEQ ID NO: 945)



CCGACUUCAUACCUUGGUAACUGAA







(SEQ ID NO: 946)



CCAGACAUGACUUGGGACAUCAAAU







(SEQ ID NO: 947)



CAGACAUGACUUGGGACAUCAAAUA







(SEQ ID NO: 948)



GACAUGACUUGGGACAUCAAAUAUA







MRPS18B



(SEQ ID NO: 949)



GCGUCUGUAUUAAACACCGUGCUGA







(SEQ ID NO: 950)



GGCGGCUUCCUAUGCUAUCUCUCUU







(SEQ ID NO: 951)



CCUCAGUUCCCAUUUCUCCUUAUAA







(SEQ ID NO: 952)



GCCCUGGAAAUAUCUGGAAUCAGAA







(SEQ ID NO: 953)



CGGAAUAAAGUUGUUGGGAAUCCCU







MTHFD1



(SEQ ID NO: 954)



CACUCACAUUAAGUUACCAAGAACA







(SEQ ID NO: 955)



CAGCUACCUUUAGAUUCAGAGAAUU







(SEQ ID NO: 956)



CCAUCUGGAUGAGGAGGUAAAUAAA







(SEQ ID NO: 957)



CAGGAAAGUGGAUGAUUCAGUAUAA







(SEQ ID NO: 958)



CAAAGUUCUGCUGUCAGCACUAGAA







MTMR11



(SEQ ID NO: 959)



GAAGGGCCUGGAACCAGAAUUGUCU







(SEQ ID NO: 960)



CCAGAAUUGUCUGGAACCCUGAUCU







(SEQ ID NO: 961)



GAAUUGUCUGGAACCCUGAUCUGUA







(SEQ ID NO: 962)



GGAACCCUGAUCUGUACCAACUUUA







(SEQ ID NO: 963)



CCCUGGUCAACAUUGGACGAUUAGA







MYC



(SEQ ID NO: 964)



CCAACAGGAACUAUGACCUCGACUA







(SEQ ID NO: 965)



GGAGGAGACAUGGUGAACCAGAGUU







(SEQ ID NO: 966)



GAGGAGACAUGGUGAACCAGAGUUU







(SEQ ID NO: 967)



GGAGACAUGGUGAACCAGAGUUUCA







(SEQ ID NO: 968)



GAGACAUGGUGAACCAGAGUUUCAU







NAT13



(SEQ ID NO: 969)



GCUGGGAGAUGUGACACCACACAAU







(SEQ ID NO: 970)



CAGGUCAUCUUUCCAGUCAGCUACA







(SEQ ID NO: 971)



GGGUGGAUCAUUCACAGAAUCAGAA







(SEQ ID NO: 972)



UGACACUAGGAUGUCUGGCACCUUA







(SEQ ID NO: 973)



CCGAAGGCUAGGAAUAGGAACUAAA







NCRNA00099



(SEQ ID NO: 974)



CAAGGGAUCUGUGAUUGUAUGUUAA







(SEQ ID NO: 975)



CAGUUGCAUGCAGAGUGAGUUUGAA







(SEQ ID NO: 976)



GGGCCAUGCUUUAUUGACAAUAUUU







(SEQ ID NO: 977)



CCACAUUAUUUAGCUCUCCCUUAUU







(SEQ ID NO: 978)



CCCUGACUGAUAGCUUUCAUCUGUA







NDUFB3



(SEQ ID NO: 979)



ACAUGGACAUGAGCAUGGACAUCAU







(SEQ ID NO: 980)



CAUGGACAUGAGCAUGGACAUCAUA







(SEQ ID NO: 981)



UGGACAUGAGCAUGGACAUCAUAAA







(SEQ ID NO: 982)



GAACUUCCAGAUUAUAGACAAUGGA







(SEQ ID NO: 983)



CCAGAUUAUAGACAAUGGAAGAUAG







NDUFS5



(SEQ ID NO: 984)



GGUUCGGCCUUAACAUAGAUCGAUG







(SEQ ID NO: 985)



UCGGCCUUAACAUAGAUCGAUGGUU







(SEQ ID NO: 986)



CGGCCUUAACAUAGAUCGAUGGUUG







(SEQ ID NO: 987)



GGCCUUAACAUAGAUCGAUGGUUGA







(SEQ ID NO: 988)



ACAUAGAUCGAUGGUUGACAAUCCA







NFYA



(SEQ ID NO: 989)



GAGCAGUAUACAGCAAACAGCAAUA







(SEQ ID NO: 990)



CAGUAUACAGCAAACAGCAAUAGUU







(SEQ ID NO: 991)



CAAACAGCAAUAGUUCGACAGAGCA







(SEQ ID NO: 992)



AGGUAGUCCAAGGGCAGCCAUUAAU







(SEQ ID NO: 993)



GAACACAGGGUUUGCAGCAAAUACA







NPEPPS



(SEQ ID NO: 994)



CACAGAGGAUCUCUGGGAAAGUUUA







(SEQ ID NO: 995)



GAACAGGUAGAAGAUGACAGAUUAU







(SEQ ID NO: 996)



CAGUACAGCUCUGCCAUGCUGGAAA







(SEQ ID NO: 997)



CAGCUCUGCCAUGCUGGAAAGUUUA







(SEQ ID NO: 998)



GAGCUGGAAUCAUUAGCACUGUAGA







NQO1



(SEQ ID NO: 999)



UCGGACCUCUAUGCCAUGAACUUCA







(SEQ ID NO: 1000)



UCGGACCUCUAUGCCAUGAACUUCA







(SEQ ID NO: 1001)



GAAAGGACAUCACAGGUAAACUGAA







(SEQ ID NO: 1002)



UGCCGAGUCUGUUCUGGCUUAUAAA







(SEQ ID NO: 1003)



CAGGUAAACUGAAGGACCCUGCGAA







(SEQ ID NO: 1004)



CGAGUCUGUUCUGGCUUAUAAAGAA







NUAK2



(SEQ ID NO: 1005)



CCCUAAUGAAGAAGCAGGCGGUGAA







(SEQ ID NO: 1006)



CGGAGGGAGAUUGAGAUCAUGUCAU







(SEQ ID NO: 1007)



CAGAUCGUCUCUGCCGUGCACUAUU







(SEQ ID NO: 1008)



CCUCGCCAGAGAUUGUCAAUGGGAA







(SEQ ID NO: 1009)



CAGAGAUUGUCAAUGGGAAGCCCUA







PAX8



(SEQ ID NO: 1010)



CAAACGCCAGAACCCUACCAUGUUU







(SEQ ID NO: 1011)



ACUGUGCCCAGUGUCAGCUCCAUUA







(SEQ ID NO: 1012)



UCCGGACCAAAGUGCAGCAACCAUU







(SEQ ID NO: 1013)



GGACCAAAGUGCAGCAACCAUUCAA







(SEQ ID NO: 1014)



CAGUGAUCAGGAUAGCUGCCGACUA







PBLD



(SEQ ID NO: 1015)



CCUGCUGCUGUUUGCCUCCUAGAAA







(SEQ ID NO: 1016)



GGAGAUGAACCUCUCUGAAACUGCU







(SEQ ID NO: 1017)



CAACUUUGCACAAAGUUCCUGCUUU







(SEQ ID NO: 1018)



UCCUGCUUUGGACUGAGAUGGUUUA







(SEQ ID NO: 1019)



GCAUCGUCCUGGACUUGCCUCUUUA







PCBP2



(SEQ ID NO: 1020)



CCGGUGUGAUUGAAGGUGGAUUAAA







(SEQ ID NO: 1021)



CGGUGUGAUUGAAGGUGGAUUAAAU







(SEQ ID NO: 1022)



GAAGGUGGAUUAAAUGUCACUCUCA







(SEQ ID NO: 1023)



UCACUCUCACCAUCCGGCUACUUAU







(SEQ ID NO: 1024)



CAUCGGAAAGAAAGGAGAAUCAGUU







PDCD10



(SEQ ID NO: 1025)



UCUAUGCAGUCAUGUAUCCUGUGUU







(SEQ ID NO: 1026)



CAGACACUGAGAGCCGCUUUCAUCA







(SEQ ID NO: 1027)



CCUUCUUCGUAUGGCAGCUGAUGAU







(SEQ ID NO: 1028)



CAGAGCCAGAAUUCCAAGACCUAAA







(SEQ ID NO: 1029)



CCAGAAUUCCAAGACCUAAACGAAA







PKP1



(SEQ ID NO: 1030)



CCACUCCAAUCGAGGUUCCAUGUAU







(SEQ ID NO: 1031)



CACUCCAAUCGAGGUUCCAUGUAUG







(SEQ ID NO: 1032)



CCAUGUAUGAUGGCUUGGCUGACAA







(SEQ ID NO: 1033)



GCAGCGACAUCUGCUUCAUGCAGAA







(SEQ ID NO: 1034)



UCUUGUUCCGCAAAGGGCCACUAGU







PLCB3



(SEQ ID NO: 1035)



CCGGUGAACACAGUGUUCUUGAACU







(SEQ ID NO: 1036)



AGGUCUGGUCUGAGGAGCUAUUCAA







(SEQ ID NO: 1037)



AGGAGCUAUUCAAGCUGGCUAUGAA







(SEQ ID NO: 1038)



CCGUCAAGAACAUCCUGAAGAUGUU







(SEQ ID NO: 1039)



CAGCCACUGAGUGCCUACUUCAUCA







POLH



(SEQ ID NO: 1040)



CACUAGAAGUAUGUGGGCAGAUGAU







(SEQ ID NO: 1041)



CAGCAUUGAUGAGGCUUACGUAGAU







(SEQ ID NO: 1042)



CCUAUCUCGGCAGACUUGUUGCCAA







(SEQ ID NO: 1043)



CAGCCAUAGAGAGGGAGACUGGUUU







(SEQ ID NO: 1044)



CAGUUAAACCCAGGCAACUACCCAA







PPP1R10



(SEQ ID NO: 1045)



AGACACCAUCCUUGGUGCCUGUGAA







(SEQ ID NO: 1046)



CAGAGAAGAAAUACAAGCCACUCAA







(SEQ ID NO: 1047)



CAAGCCACUCAACACAACACCUAAU







(SEQ ID NO: 1048)



CCAAAGAGAUCAAAGUGAAGAUCAU







(SEQ ID NO: 1049)



CCUGGGCUUUCUGGAUGCUCUUAAU







PTRH2



(SEQ ID NO: 1050)



CAAGACGAGCAAGACACACACAGAU







(SEQ ID NO: 1051)



GAGCAAGACACACACAGAUACUGAA







(SEQ ID NO: 1052)



CCAGUGCUCUCAUGCUGCUGUUUCA







(SEQ ID NO: 1053)



GCUGCUGUUUCAGCCUACAAGCAGA







(SEQ ID NO: 1054)



CAAGGUGGUGGUCAAAGCUCCUGAU







PXN



(SEQ ID NO: 1055)



UCCGACUUUGAUAGAUUUCUA







(SEQ ID NO: 1056)



CCGACUGAAACUGGAACCCUU







(SEQ ID NO: 1057)



CCUGUGAUUUAUGCCAAUAAA







(SEQ ID NO: 1058)



CUGCUGGAACUGAACGCUGUA







(SEQ ID NO: 1059)



UACACUGAGGUUUGAAUUCAU







QARS



(SEQ ID NO: 1060)



GAGGCUGAUCUGGAGAAGAAGUUCA







(SEQ ID NO: 1061)



CACACACCAUGAAUCUACUAAAGCA







(SEQ ID NO: 1062)



CAGAACCCAAUGGAAUCCUGCAUAU







(SEQ ID NO: 1063)



GGACAUGCCAAAGCCAUCAAUUUCA







(SEQ ID NO: 1064)



CCUGUAGCCUAUCGAGUCAAGUAUA







RABL3



(SEQ ID NO: 1065)



UGGAUCGGGUGAAGGUACUGGUGUU







(SEQ ID NO: 1066)



GGAGACUCAGGUGUUGGGAAAUCUU







(SEQ ID NO: 1067)



GCUGCUCAGUGGAUGUCAGAGUUCA







(SEQ ID NO: 1068)



CAGUGGAUGUCAGAGUUCAUGAUUA







(SEQ ID NO: 1069)



CCCAGAAGAGAAGACCUACUACAUA







RAD51L1



(SEQ ID NO: 1070)



CAAGAGCUGUGUGACCGUCUGAGUA







(SEQ ID NO: 1071)



UGGAGCUUAUGAAGGUGACUGGUCU







(SEQ ID NO: 1072)



GAGGUGUCCAUGAACUUCUAUGUAU







(SEQ ID NO: 1073)



GAAGGAGCUGUGGUGUACAUUGACA







(SEQ ID NO: 1074)



GGGAACUCACCUGUGAUGAAGUUCU







RBMX



(SEQ ID NO: 1075)



CGCUGCCCUCUCGUAGAGAUGUUUA







(SEQ ID NO: 1076)



UGCCCUCUCGUAGAGAUGUUUAUUU







(SEQ ID NO: 1077)



CCCUCUCGUAGAGAUGUUUAUUUGU







(SEQ ID NO: 1078)



CCCAAGAGAUGAUGGGUAUUCUACU







(SEQ ID NO: 1079)



GGGUAUUCUACUAAAGACAGCUAUU







RGD1308059



(SEQ ID NO: 1080)



CCAGAUCUCAGAGACAUCGAGCUGA







(SEQ ID NO: 1081)



CAGAUCUCAGAGACAUCGAGCUGAA







(SEQ ID NO: 1082)



GGCUAGAGACCAAACUGCACAUCCU







(SEQ ID NO: 1083)



ACGUAAGGCUCAUGCGUCAAUUGCU







(SEQ ID NO: 1084)



GGCUCAUGCGUCAAUUGCUCCUCAU







RGD1309079



(SEQ ID NO: 1085)



AGAGUUGACCUUGCUUGGCAGUUAA







(SEQ ID NO: 1086)



GAGUUGACCUUGCUUGGCAGUUAAU







(SEQ ID NO: 1087)



GGAAGGUAAUCAUUGUGGAUGCUGA







(SEQ ID NO: 1088)



GAUGUAUGGUAUGAGAGCAAGUUUG







(SEQ ID NO: 1089)



UGACUGUGACAAGGAGGCUUGCUUA







RGD1309492



(SEQ ID NO: 1090)



CAGCCCUCCUUUGAAGUGAUCUCAG







(SEQ ID NO: 1091)



AGUCCAGAAACUACUUGCCCAUUCU







(SEQ ID NO: 1092)



CCAGAAACUACUUGCCCAUUCUAAA







(SEQ ID NO: 1093)



CAGAAACUACUUGCCCAUUCUAAAU







(SEQ ID NO: 1094)



CAUCAGACAGCACUCUGCAUGGCUU







RMI1



(SEQ ID NO: 1095)



CAUGGCAUGUUAAAGUACCUCCGAU







(SEQ ID NO: 1096)



UCCGAUGUGGCUGGAAGCUUGUAUU







(SEQ ID NO: 1097)



AGGCCCAAAUGAAUAAACAAGUGUU







(SEQ ID NO: 1098)



UGGUUGAUGUAAGUCAGCCUGCAUA







(SEQ ID NO: 1099)



CCAGAAAGAACAGAUGGAAACUAAG







RPL18AP3



(SEQ ID NO: 1070)



CCGCAUGCGAAUCUUUGCGCCUAAU







(SEQ ID NO: 1071)



GCAUGCGAAUCUUUGCGCCUAAUCA







(SEQ ID NO: 1072)



CAUGCGAAUCUUUGCGCCUAAUCAU







(SEQ ID NO: 1073)



CAAGUCCCGCUUCUGGUACUUUGUA







(SEQ ID NO: 1074)



CCCGCUUCUGGUACUUUGUAUCUCA







RPL27A



(SEQ ID NO: 1075)



GCGGUAAUGCUGGUGGUCUGCAUCA







(SEQ ID NO: 1076)



CAUCACCACCGGAUCAACUUCGACA







(SEQ ID NO: 1077)



CAGGCUACUUUGGGAAAGUUGGUAU







(SEQ ID NO: 1078)



ACAGACACGGGUGAAUGCUGCUAAA







(SEQ ID NO: 1079)



GAUGUGGUGCGAUCGGGCUACUACA







RPL29P2



(SEQ ID NO: 1080)



CCCUACAAAGACUUCAGAGUAGAUA







(SEQ ID NO: 1081)



CCUACAAAGACUUCAGAGUAGAUAU







(SEQ ID NO: 1082)



CAAAGACUUCAGAGUAGAUAUCUCU







(SEQ ID NO: 1083)



CCUCCUGCGCUAUUGGUACAAAUAA







(SEQ ID NO: 1084)



GCGCUAUUGGUACAAAUAAGCCUGA







RPL29P31



(SEQ ID NO: 1085)



CCCGAUCACGAAGAUAUGAAUCUCU







(SEQ ID NO: 1086)



CCGAUCACGAAGAUAUGAAUCUCUU







(SEQ ID NO: 1087)



CGAUCACGAAGAUAUGAAUCUCUUA







RPL3



(SEQ ID NO: 1088)



CCGGACCUUCAAGACUGUCUUUGCU







(SEQ ID NO: 1089)



GAUGAAUGCAAGAGGCGUUUCUAUA







(SEQ ID NO: 1090)



CAGGUACCUGUGAACCAAGUGUUUG







(SEQ ID NO: 1091)



ACCAAGUGUUUGGGCAGGAUGAGAU







(SEQ ID NO: 1092)



CGCACUGAGAUCAACAAGAAGAUUU







RPL34



(SEQ ID NO: 1093)



AGAUACUACUCAAGUUACAGUAUAU







(SEQ ID NO: 1094)



CAAGUUACAGUAUAUGAUCACUAAU







(SEQ ID NO: 1095)



CAGGAGCUCUGAUAUAUAUCUGGUA







(SEQ ID NO: 1096)



GGAGCUCUGAUAUAUAUCUGGUACA







(SEQ ID NO: 1097)



GCUCUGAUAUAUAUCUGGUACAUGU







RPL37A



(SEQ ID NO: 1098)



CCAAGAAAGUCGGGAUCGUCGGUAA







(SEQ ID NO: 1099)



CAAGAAAGUCGGGAUCGUCGGUAAA







(SEQ ID NO: 1100)



CGUCGGUAAAUACGGGACCCGCUAU







(SEQ ID NO: 1101)



AAAUUGAAAUCAGCCAGCACGCCAA







(SEQ ID NO: 1102)



GAAAUCAGCCAGCACGCCAAGUACA







RPL4



(SEQ ID NO: 1103)



CCACUGAUAUCGGUGUACUCCGAAA







(SEQ ID NO: 1104)



CAGACAGCCCUAUGCUGUCAGUGAA







(SEQ ID NO: 1105)



GCCCUACCAGCACUGGUCAUGUCUA







(SEQ ID NO: 1106)



UGGUCAUGUCUAAAGGUCAUCGUAU







(SEQ ID NO: 1107)



GGAAGUUCCUGAACUUCCUUUGGUA







RPL7A



(SEQ ID NO: 1108)



CCGAAAGGAAAGAAGGCCAAGGGAA







(SEQ ID NO: 1109)



CGUGAAGAAGCAGGAGGCUAAGAAA







(SEQ ID NO: 1110)



CCCGCUAUAUCAGGUUGCAGCGGCA







(SEQ ID NO: 1111)



CAGCGGCAGAGAGCCAUCCUCUAUA







(SEQ ID NO: 1112)



GAGCCAUCCUCUAUAAGCGGCUGAA







RPS18



(SEQ ID NO: 1113)



UGGAACGUGUGAUCACCAUUA







(SEQ ID NO: 1114)



AAGAAGAUUCGGGCCCAUAGA







(SEQ ID NO: 1115)



CUGGACAACAAGCUCCGUGAA







(SEQ ID NO: 1116)



ACGCCAGUACAAGAUCCCAGA







RPSA



(SEQ ID NO: 1117)



AAGUUGUCUUCAUUUAGUUUGCUUU







(SEQ ID NO: 1118)



UACUCCAGAUCAGAAUACCUGGGAU







(SEQ ID NO: 1119)



ACUCCAGAUCAGAAUACCUGGGAUU







(SEQ ID NO: 1120)



CCAGAUCAGAAUACCUGGGAUUGCA







(SEQ ID NO: 1121)



CAGAUCAGAAUACCUGGGAUUGCAU







SDF2



(SEQ ID NO: 1122)



UGGCUGUAGUACCUCUGCUGUUGUU







(SEQ ID NO: 1123)



GCCACAACGUCCGACUGCACUCACA







(SEQ ID NO: 1124)



ACCUCUGUGGAUGACAGCAACAGUU







(SEQ ID NO: 1125)



UGGAUGACUGGACAGUGCUCUGUAA







(SEQ ID NO: 1126)



GAGAGAUGGUGAGGUGCGGUUCAAA







SEMA3C



(SEQ ID NO: 1127)



GGGAGGAGAUCAGAGGACCAAGUUU







(SEQ ID NO: 1128)



CCAAAUGGCUAAGUGAACCUAUGUU







(SEQ ID NO: 1129)



CAUCCCAGAUGGUACUGAUCCAAAU







(SEQ ID NO: 1130)



GAUCAGCCGUGUGUGUGUAUCAUUU







(SEQ ID NO: 1131)



CGUAUUGGCACUGACUACAAGUAUA







SERAC1



(SEQ ID NO: 1132)



CGGAGACCCAUCACUGGCAUGAUUA







(SEQ ID NO: 1133)



CAUCACUGGCAUGAUUACCAGUAUA







(SEQ ID NO: 1134)



CCUACCACCUCCUUUGCCAUCUUUA







(SEQ ID NO: 1135)



GCUCAGACAGUUGCUGGCUUCCUUA







(SEQ ID NO: 1136)



GGACAGUGAGCAGGCUGUAAUUGAA







SERPINI1



(SEQ ID NO: 1137)



GCUGACUUGUCAGUGAAUAUGUAUA







(SEQ ID NO: 1138)



CACUCAAUGGGAUAUGACAGCCUAA







(SEQ ID NO: 1139)



GCCCUCAUUAAUGCUGUCUAUUUCA







(SEQ ID NO: 1140)



CCAAGUCCUAGAAAUACCAUAUGAA







(SEQ ID NO: 1141)



UGGCUGUGCUGUAUCCUCAAGUUAU







SF3B4



(SEQ ID NO: 1142)



GGGCCUGGAUGAGAAGGUUAGUGAA







(SEQ ID NO: 1143)



AAGGCUAUGGCUUUGUGGAAUUCUU







(SEQ ID NO: 1144)



GGCUAUGGCUUUGUGGAAUUCUUGA







(SEQ ID NO: 1145)



GAGGAAGAUGCUGACUAUGCCAUUA







(SEQ ID NO: 1146)



UGAACAUGAUCAAACUCUAUGGGAA







SFRS3



(SEQ ID NO: 1147)



AACCCUAGAUCUCGAAAUGCA







(SEQ ID NO: 1148)



AAGCUGAUCCAUCAUGAUGUA







(SEQ ID NO: 1149)



GAGCUAGAUGGAAGAACACUA







(SEQ ID NO: 1150)



CUCGUAGUCGAUCUAGGUCAA







SFXN1



(SEQ ID NO: 1151)



CUCGAUGGGAUCAAAGCACUU







(SEQ ID NO: 1152)



AGGACUCAAUGCAUUGACCAA







(SEQ ID NO: 1153)



CGCGUGUACUUCAAUAAGGGA







(SEQ ID NO: 1154)



UCGGCGAACGCUGCGAAACAA







SKIL



(SEQ ID NO: 1155)



CCAAGAAAGCAUGUCGCCUACUGUA







(SEQ ID NO: 1156)



CCACAGAACUCACUCAGACUGUGUU







(SEQ ID NO: 1157)



CAGUGUGUGAUGAACUGUACAUAUA







(SEQ ID NO: 1158)



GGUGUACUUCAGACCAGCUUCAUAU







(SEQ ID NO: 1159)



AGGUACUGGGCAUACUUCCAUUCAA







SLC25A25



(SEQ ID NO: 1160)



CGGGACUUGGGAGUCAAGAUAUCUG







(SEQ ID NO: 1161)



CCACGAUCUUUGAUGUGGGUGAGAA







(SEQ ID NO: 1162)



CACGAUCUUUGAUGUGGGUGAGAAU







(SEQ ID NO: 1163)



GAUCUUUGAUGUGGGUGAGAAUCUA







(SEQ ID NO: 1164)



GCAUCGUUGGUGGCUUCACUCAGAU







SLC38A2



(SEQ ID NO: 1165)



CAUCCAGGUACUACUUCCUUUGGAA







(SEQ ID NO: 1166)



CAGUGGAAUCCUUGGGCUUUCUUAU







(SEQ ID NO: 1167)



GACUGCCAAUGAAGGAGGGUCUUUA







(SEQ ID NO: 1168)



GCCAAUGAAGGAGGGUCUUUAUUAU







(SEQ ID NO: 1169)



GCUACCUCUUCAUAGUGAAAUAUGA







SLC39A14



(SEQ ID NO: 1170)



CAAGAGGCUGCUGCUCUACUUCAUA







(SEQ ID NO: 1171)



CGGAGGCAUUUGGUUUCAACCCUCU







(SEQ ID NO: 1172)



GAAGAUUAUUAUGUCUCCAAGUCUG







(SEQ ID NO: 1173)



CAGAGAAGAUCUUGAAGAUUCUUCU







(SEQ ID NO: 1174)



CAUCAUCAUGGACACAGCCAUUAUG







SMC6



(SEQ ID NO: 1175)



GAGGCAUAGAGACAGUGCUACUAAU







(SEQ ID NO: 1176)



UGCAGGACGUUAUUAUUCAUCUGAA







(SEQ ID NO: 1177)



UCAGCAACAUUUAUCUGCCCUUGAA







(SEQ ID NO: 1178)



CAGCAACAUUUAUCUGCCCUUGAAA







(SEQ ID NO: 1179)



CCAGUCUGUAGAUAUUGCAACUUUG







SNORA1



(SEQ ID NO: 1180)



GAAUGGGCACUGUUGAUCAUGGUGU







(SEQ ID NO: 1181)



UGGGCACUGUUGAUCAUGGUGUCCA







(SEQ ID NO: 1182)



GGGCACUGUUGAUCAUGGUGUCCAA







(SEQ ID NO: 1183)



UGGCUAAAUUGAGACAGGUUAUGCU







(SEQ ID NO: 1184)



UGAGACAGGUUAUGCUUCCAUCACA







SNORA18



(SEQ ID NO: 1185)



CCUGUAGCCUGCACAUCGUUGGAAA







(SEQ ID NO: 1186)



UGCACAUCGUUGGAAACGCCUCAUA







(SEQ ID NO: 1187)



GCACAUCGUUGGAAACGCCUCAUAG







(SEQ ID NO: 1188)



GGAAACGCCUCAUAGAGUAACUCUG







(SEQ ID NO: 1189)



ACGCCUCAUAGAGUAACUCUGUGGU







SNORA19



(SEQ ID NO: 1190)



GCACAUUUCAUUGACCUGCUUUCUU







(SEQ ID NO: 1191)



CACAUUUCAUUGACCUGCUUUCUUU







(SEQ ID NO: 1192)



GAGUAGUGUUAUUUCUUAUGUGCUA







(SEQ ID NO: 1193)



UAUUUCUUAUGUGCUAUACAAAUAA







(SEQ ID NO: 1194)



GCUAUACAAAUAAUUGAAGGCUAAU







SNORA3



(SEQ ID NO: 1195)



UCGAGGCUAGAGUCACGCUUGGGUA







(SEQ ID NO: 1196)



GAGUCACGCUUGGGUAUCGGCUAUU







(SEQ ID NO: 1197)



GCUAUUGCCUGAGUGUGCUAGAGUC







(SEQ ID NO: 1198)



GAGUGUGCUAGAGUCCUCGAAGAGU







(SEQ ID NO: 1199)



CCUCGAAGAGUAACUGCUGACCUUA







SNORA32



(SEQ ID NO: 1200)



UGCAGUUUCUCAUUUGCUGUGGACA







(SEQ ID NO: 1201)



UCAUUUGCUGUGGACAUGACCAUAA







(SEQ ID NO: 1202)



CAUUUGCUGUGGACAUGACCAUAAA







(SEQ ID NO: 1203)



UGCUACUUUGCUAGCAAUCAGCUUA







(SEQ ID NO: 1204)



GCUACUUUGCUAGCAAUCAGCUUAU







SNORA38



(SEQ ID NO: 1205)



CCUCCUACAAAGGCGUGUCUGUGGU







(SEQ ID NO: 1206)



AGGCGUGUCUGUGGUUCCCUGUCUU







(SEQ ID NO: 1207)



GGCGUGUCUGUGGUUCCCUGUCUUU







(SEQ ID NO: 1208)



UGGUUCCCUGUCUUUGGACACGUAA







(SEQ ID NO: 1209)



CCCUGUCUUUGGACACGUAAGAAUU







SNORA40



(SEQ ID NO: 1210)



CAACCCAGAACUCAUUGUUCAGUAU







(SEQ ID NO: 1211)



CCCAGAACUCAUUGUUCAGUAUGAG







(SEQ ID NO: 1212)



CCAGAACUCAUUGUUCAGUAUGAGU







(SEQ ID NO: 1213)



CAGAACUCAUUGUUCAGUAUGAGUU







(SEQ ID NO: 1214)



AGAACUCAUUGUUCAGUAUGAGUUU







SNORA45



(SEQ ID NO: 1215)



CCUGACACAACUCUUGUCCUGGUGU







(SEQ ID NO: 1216)



GACACAACUCUUGUCCUGGUGUGCU







(SEQ ID NO: 1217)



ACUCUUGUCCUGGUGUGCUAGAGUA







(SEQ ID NO: 1218)



CCUGGUGUGCUAGAGUACUCGAAGA







(SEQ ID NO: 1219)



GGUGUGCUAGAGUACUCGAAGAGAA







SNORA54



(SEQ ID NO: 1220)



ACCCGUUAGCCUGGCUGUAGCUAAU







(SEQ ID NO: 1221)



UAAUGGGUUCCAUUCCGGUGCAAUA







(SEQ ID NO: 1222)



GGUUCCAUUCCGGUGCAAUAGCAUU







(SEQ ID NO: 1223)



CCAUUCCGGUGCAAUAGCAUUUCCA







(SEQ ID NO: 1224)



CGGUGCAAUAGCAUUUCCAGCGACA







SNORA6



(SEQ ID NO: 1225)



GCACACUAUUAAAGCUCAGGGUGGA







(SEQ ID NO: 1226)



AAAGCUCAGGGUGGAGGCCAGUCUU







(SEQ ID NO: 1227)



GGAGGCCAGUCUUGGCUCAUGAACU







(SEQ ID NO: 1228)



GAGGCCAGUCUUGGCUCAUGAACUU







(SEQ ID NO: 1229)



GGCCAGUCUUGGCUCAUGAACUUCU







SNORA62



(SEQ ID NO: 1230)



GCACAUUGUUAGAGCUUGGAGUUGA







(SEQ ID NO: 1231)



CACAUUGUUAGAGCUUGGAGUUGAG







(SEQ ID NO: 1232)



GAGCUUGGAGUUGAGGCUACUGACU







(SEQ ID NO: 1233)



ACUGGCCGAUGAACUCGCAAGUGUA







(SEQ ID NO: 1234)



GCCCAAUGAGUGGAGUUUGAUAGUA







SNORA76



(SEQ ID NO: 1235)



GGUCAAUCUGCAGCGCUAGAGCAUG







(SEQ ID NO: 1236)



CAGCGCUAGAGCAUGUGCUUGCGCA







(SEQ ID NO: 1237)



GCGCUAGAGCAUGUGCUUGCGCAUA







(SEQ ID NO: 1238)



CGCUAGAGCAUGUGCUUGCGCAUAA







(SEQ ID NO: 1239)



GCUAGAGCAUGUGCUUGCGCAUAAC







SNORA8



(SEQ ID NO: 1240)



CACUGCAUGGUAUCUGCACUCAGCA







(SEQ ID NO: 1241)



CAUGGUAUCUGCACUCAGCAGUUUA







(SEQ ID NO: 1242)



CAGUUUACACCUGCUAGGGUGUUCA







(SEQ ID NO: 1243)



CACCUGCUAGGGUGUUCAAAGGUCA







(SEQ ID NO: 1244)



CAAAGGUCAGUGCUAUAGAAAUUCA







SNORA84



(SEQ ID NO: 1245)



CCUGUGGUUGCUGGAUGCUGUUGUG







(SEQ ID NO: 1246)



UGCUGGAUGCUGUUGUGCAUGGACA







(SEQ ID NO: 1247)



GAUGCUGUUGUGCAUGGACAGCUCU







(SEQ ID NO: 1248)



CCAGUGGAUUCGAUGGGCCAUAGCA







(SEQ ID NO: 1249)



CAGUGGAUUCGAUGGGCCAUAGCAA







SNORD16



(SEQ ID NO: 1250)



CAAUGAUGUCGUAAUUUGCGUCUUA







(SEQ ID NO: 1251)



GAUGUCGUAAUUUGCGUCUUACUCU







(SEQ ID NO: 1252)



CGUAAUUUGCGUCUUACUCUGUUCU







(SEQ ID NO: 1253)



GCGUCUUACUCUGUUCUCAGCGACA







(SEQ ID NO: 1254)



UCUUACUCUGUUCUCAGCGACAGUU







SNORD18A



(SEQ ID NO: 1255)



CAGUAGUGAUGAAAUUCCACUUCAU







(SEQ ID NO: 1256)



AGUAGUGAUGAAAUUCCACUUCAUU







(SEQ ID NO: 1257)



GAAAUUCCACUUCAUUGGUCCGUGU







(SEQ ID NO: 1258)



AAAUUCCACUUCAUUGGUCCGUGUU







(SEQ ID NO: 1259)



AAUUCCACUUCAUUGGUCCGUGUUU







SNORD18B



(SEQ ID NO: 1260)



GAGAUUCCACUUAAUUGGUCCGUGU







(SEQ ID NO: 1261)



AGAUUCCACUUAAUUGGUCCGUGUU







(SEQ ID NO: 1262)



GAUUCCACUUAAUUGGUCCGUGUUU







(SEQ ID NO: 1263)



CCACUUAAUUGGUCCGUGUUUCUGA







(SEQ ID NO: 1264)



CACUUAAUUGGUCCGUGUUUCUGAA







SNORD18C



(SEQ ID NO: 1265)



UGAGAUUCCACUUAAGGUCCGUGUU







(SEQ ID NO: 1266)



GAGAUUCCACUUAAGGUCCGUGUUU







(SEQ ID NO: 1267)



GAUUCCACUUAAGGUCCGUGUUUCU







(SEQ ID NO: 1268)



UCCACUUAAGGUCCGUGUUUCUGAA







(SEQ ID NO: 1269)



ACUUAAGGUCCGUGUUUCUGAAACA







SNORD24



(SEQ ID NO: 1270)



GAAUAUUUGCUAUCUGAGAGAUGGU







(SEQ ID NO: 1271)



UAUUUGCUAUCUGAGAGAUGGUGAU







(SEQ ID NO: 1272)



UGCUAUCUGAGAGAUGGUGAUGACA







(SEQ ID NO: 1273)



GCUAUCUGAGAGAUGGUGAUGACAU







(SEQ ID NO: 1274)



UAUCUGAGAGAUGGUGAUGACAUUU







SNORD35B



(SEQ ID NO: 1275)



UGGUCUUCAGAUGCCCACGUGGGCA







(SEQ ID NO: 1276)



AGAUGCCCACGUGGGCACUGCUGAG







(SEQ ID NO: 1277)



GAUGCCCACGUGGGCACUGCUGAGA







(SEQ ID NO: 1278)



UGCCCACGUGGGCACUGCUGAGAAA







(SEQ ID NO: 1279)



CACGUGGGCACUGCUGAGAAAGCCA







SNORD36A



(SEQ ID NO: 1280)



GCAAUGAUGUGAAUCUCUCACUGAA







(SEQ ID NO: 1281)



CAAUGAUGUGAAUCUCUCACUGAAU







(SEQ ID NO: 1282)



AAUGAUGUGAAUCUCUCACUGAAUU







(SEQ ID NO: 1283)



UGAUGUGAAUCUCUCACUGAAUUCA







(SEQ ID NO: 1284)



GAUGUGAAUCUCUCACUGAAUUCAA







SNORD36B



(SEQ ID NO: 1285)



UCUUGGCCUGAAAUUACUGUGAAGA







(SEQ ID NO: 1286)



UGGCCUGAAAUUACUGUGAAGAGUA







(SEQ ID NO: 1287)



GGCCUGAAAUUACUGUGAAGAGUAA







(SEQ ID NO: 1288)



GCCUGAAAUUACUGUGAAGAGUAAA







SNORD36C



(SEQ ID NO: 1289)



UGCCAAUGAUGGUUAAGAAUUUCUU







(SEQ ID NO: 1290)



CCAAUGAUGGUUAAGAAUUUCUUCA







(SEQ ID NO: 1291)



GAUGGUUAAGAAUUUCUUCACCUGA







(SEQ ID NO: 1292)



UGGUUAAGAAUUUCUUCACCUGAAU







(SEQ ID NO: 1293)



GGUUAAGAAUUUCUUCACCUGAAUA







SNORD3B-2



(SEQ ID NO: 1294)



AAGACUAUACUUUCAGGGAUCAUUU







(SEQ ID NO: 1295)



ACUAUACUUUCAGGGAUCAUUUCUA







(SEQ ID NO: 1296)



CAGGGAUCAUUUCUAUAGUGUGUUA







(SEQ ID NO: 1297)



GGGAUCAUUUCUAUAGUGUGUUACU







(SEQ ID NO: 1298)



GAAGUUUCUCUGAACGUGUAGAGCA







SNORD43



(SEQ ID NO: 1299)



GAUGAUGAACUUAUUGACGGGCGGA







(SEQ ID NO: 1300)



GAUGAACUUAUUGACGGGCGGACAG







(SEQ ID NO: 1301)



UGAACUUAUUGACGGGCGGACAGAA







(SEQ ID NO: 1302)



GAACUUAUUGACGGGCGGACAGAAA







(SEQ ID NO: 1303)



GACGGGCGGACAGAAACUGUGUGCU







SNORD44



(SEQ ID NO: 1304)



CCUGGAUGAUGAUAAGCAAAUGCUG







(SEQ ID NO: 1305)



UGAUGAUAAGCAAAUGCUGACUGAA







(SEQ ID NO: 1306)



UAAGCAAAUGCUGACUGAACAUGAA







(SEQ ID NO: 1307)



AAUGCUGACUGAACAUGAAGGUCUU







(SEQ ID NO: 1308)



GCUGACUGAACAUGAAGGUCUUAAU







SNORD47



(SEQ ID NO: 1309)



ACCAAUGAUGUAAUGAUUCUGCCAA







(SEQ ID NO: 1310)



CCAAUGAUGUAAUGAUUCUGCCAAA







(SEQ ID NO: 1311)



CAAUGAUGUAAUGAUUCUGCCAAAU







(SEQ ID NO: 1312)



UGAUGUAAUGAUUCUGCCAAAUGAA







(SEQ ID NO: 1313)



GAUGUAAUGAUUCUGCCAAAUGAAA







SNORD5



(SEQ ID NO: 1314)



UCAGAUGAUGAAUUUAACUGUUCAA







(SEQ ID NO: 1315)



GAUGAUGAAUUUAACUGUUCAACUG







(SEQ ID NO: 1316)



UGAUGAAUUUAACUGUUCAACUGCU







(SEQ ID NO: 1317)



UGAAUUUAACUGUUCAACUGCUGAA







(SEQ ID NO: 1318)



UAACUGUUCAACUGCUGAAUGAUAA







SNORD58A



(SEQ ID NO: 1319)



CAGUGAUGACUUUCUUAGGACACCU







(SEQ ID NO: 1320)



AGUGAUGACUUUCUUAGGACACCUU







(SEQ ID NO: 1321)



UGAUGACUUUCUUAGGACACCUUUG







(SEQ ID NO: 1322)



UGACUUUCUUAGGACACCUUUGGAU







(SEQ ID NO: 1323)



GACUUUCUUAGGACACCUUUGGAUU







SNORD6



(SEQ ID NO: 1324)



GAUGUUAUGAUGAUGGGCGAAAUGU







(SEQ ID NO: 1325)



GAUGGGCGAAAUGUUCAACUGCUCU







(SEQ ID NO: 1326)



GGGCGAAAUGUUCAACUGCUCUGAA







(SEQ ID NO: 1327)



GGCGAAAUGUUCAACUGCUCUGAAG







(SEQ ID NO: 1328)



GCGAAAUGUUCAACUGCUCUGAAGG







SNORD60



(SEQ ID NO: 1329)



AGUCUGUGAUGAAUUGCUUUGACUU







(SEQ ID NO: 1330)



UCUGUGAUGAAUUGCUUUGACUUCU







(SEQ ID NO: 1331)



GCUUUGACUUCUGACACCUCGUAUG







(SEQ ID NO: 1332)



AACUGCACGUGCAGUCUGAUUAUUU







(SEQ ID NO: 1333)



ACUGCACGUGCAGUCUGAUUAUUUA







SNORD61



(SEQ ID NO: 1334)



UGAAUUUGAUUGCAUUGAUCGUCUG







(SEQ ID NO: 1335)



GAUUGCAUUGAUCGUCUGACAUGAU







(SEQ ID NO: 1336)



UGCAUUGAUCGUCUGACAUGAUAAU







(SEQ ID NO: 1337)



UGAUCGUCUGACAUGAUAAUGUAUU







(SEQ ID NO: 1338)



GAUCGUCUGACAUGAUAAUGUAUUU







SNORD74



(SEQ ID NO: 1339)



GCCUCUGAUGAAGCCUGUGUUGGUA







(SEQ ID NO: 1340)



GAUGAAGCCUGUGUUGGUAGGGACA







(SEQ ID NO: 1341)



GAAGCCUGUGUUGGUAGGGACAUCU







(SEQ ID NO: 1342)



GCCUGUGUUGGUAGGGACAUCUGAC







(SEQ ID NO: 1343)



CCUGUGUUGGUAGGGACAUCUGACA







SNORD75



(SEQ ID NO: 1344)



CCUGUGAUGCUUUAAGAGUAGUGGA







(SEQ ID NO: 1345)



GAUGCUUUAAGAGUAGUGGACAGAA







(SEQ ID NO: 1346)



GAGUAGUGGACAGAAGGGAUUUCUG







(SEQ ID NO: 1347)



UAGUGGACAGAAGGGAUUUCUGAAA







(SEQ ID NO: 1348)



GGACAGAAGGGAUUUCUGAAAUUCU







SNORD76



(SEQ ID NO: 1349)



UGCCACAAUGAUGACAGUUUAUUUG







(SEQ ID NO: 1350)



CCACAAUGAUGACAGUUUAUUUGCU







(SEQ ID NO: 1351)



CACAAUGAUGACAGUUUAUUUGCUA







(SEQ ID NO: 1352)



UGAUGACAGUUUAUUUGCUACUCUU







(SEQ ID NO: 1353)



GACAGUUUAUUUGCUACUCUUGAGU







SNORD77



(SEQ ID NO: 1354)



CAGAUACUAUGAUGGUUGCAUAGUU







(SEQ ID NO: 1355)



GAUACUAUGAUGGUUGCAUAGUUCA







(SEQ ID NO: 1356)



ACUAUGAUGGUUGCAUAGUUCAGCA







(SEQ ID NO: 1357)



GAUGGUUGCAUAGUUCAGCAGAUUU







(SEQ ID NO: 1358)



UGGUUGCAUAGUUCAGCAGAUUUAA







SNORD78



(SEQ ID NO: 1359)



UGUAAUGAUGUUGAUCAAAUGUCUG







(SEQ ID NO: 1360)



UGAUGUUGAUCAAAUGUCUGACCUG







(SEQ ID NO: 1361)



GAUGUUGAUCAAAUGUCUGACCUGA







(SEQ ID NO: 1362)



UGUUGAUCAAAUGUCUGACCUGAAA







(SEQ ID NO: 1363)



GAUCAAAUGUCUGACCUGAAAUGAG







SNORD80



(SEQ ID NO: 1364)



ACAAUGAUGAUAACAUAGUUCAGCA







(SEQ ID NO: 1365)



GAUGAUAACAUAGUUCAGCAGACUA







(SEQ ID NO: 1366)



CAUAGUUCAGCAGACUAACGCUGAU







(SEQ ID NO: 1367)



CAGACUAACGCUGAUGAGCAAUAUU







(SEQ ID NO: 1368)



AGACUAACGCUGAUGAGCAAUAUUA







SNORD81



(SEQ ID NO: 1369)



CAUGAUGAUCUCAAUCCAACUUGAA







(SEQ ID NO: 1370)



UGAUGAUCUCAAUCCAACUUGAACU







(SEQ ID NO: 1371)



GAUCUCAAUCCAACUUGAACUCUCU







(SEQ ID NO: 1372)



UCAAUCCAACUUGAACUCUCUCACU







(SEQ ID NO: 1373)



CAAUCCAACUUGAACUCUCUCACUG







SNORD83A



(SEQ ID NO: 1374)



GCUGUUCGUUGAUGAGGCUCAGAGU







(SEQ ID NO: 1375)



GAGCGCUGGGUACAGCGCCCGAAUC







(SEQ ID NO: 1376)



UACAGCGCCCGAAUCGGACAGUGUA







(SEQ ID NO: 1377)



CCGAAUCGGACAGUGUAGAACCAUU







(SEQ ID NO: 1378)



UCGGACAGUGUAGAACCAUUCUCUA







SNORD83B



(SEQ ID NO: 1379)



GCUGUUCAGUGAUGAGGCCUGGAAU







(SEQ ID NO: 1380)



GCCCGAGACAGACUGCGGAACCGUU







(SEQ ID NO: 1381)



GACAGACUGCGGAACCGUUCCUUGU







(SEQ ID NO: 1382)



ACAGACUGCGGAACCGUUCCUUGUU







(SEQ ID NO: 1383)



CAGACUGCGGAACCGUUCCUUGUUG







SPATA19



(SEQ ID NO: 1384)



UGAUAAUUACGACAUGGAUUGUGUA







(SEQ ID NO: 1385)



GGAUUGUGUAUAUUCUUGCUCGGAA







(SEQ ID NO: 1386)



GAUUGUGUAUAUUCUUGCUCGGAAA







(SEQ ID NO: 1387)



UGUGGAAAGUGAGGCUGUGUCUGUA







(SEQ ID NO: 1388)



CAGGGUGUAAGGGAGAAGAUGUCCA







SRP54



(SEQ ID NO: 1389)



AAUGAUUCAGCAUGCUGUAUUUAAA







(SEQ ID NO: 1390)



CAACAACAUGUUCAAAGCUAGCAUA







(SEQ ID NO: 1391)



CAACAUGUUCAAAGCUAGCAUAUUA







(SEQ ID NO: 1392)



GAGGAAAGGUUGGAAGACCUGUUUA







(SEQ ID NO: 1393)



GGAUCCUGUCAUCAUUGCUUCUGAA







ST6GAL2



(SEQ ID NO: 1394)



AAGAUGGGUUUGAACAUAAAGAGUU







(SEQ ID NO: 1395)



GGGCCUUCCUGUACCGGCUCUGGAA







(SEQ ID NO: 1396)



CGCAGCUGCGCUGUCGUCAUGUCUG







(SEQ ID NO: 1397)



CAGCCAUCACUUCAUUGACAGUUCA







(SEQ ID NO: 1398)



UCACUCCAUAUAUUCAGCAUCGUCA







STH



(SEQ ID NO: 1399)



GGGUGGAGGCCAAGUCUCAUGCAUU







(SEQ ID NO: 1401)



GGGUUAAUUUAACUCAGCCUCUGUG







(SEQ ID NO: 1402)



CAGGUUGCCAGAGACAGAACCCUCA







(SEQ ID NO: 1403)



CAGAACCCUCAGCUUAGCAUGGGAA







(SEQ ID NO: 1404)



GCAUGGGAAGUAGCUUCCCUGUUGA







SUPT6H



(SEQ ID NO: 1405)



GAAGAAGAAGCUGACUGGAUCUACA







(SEQ ID NO: 1406)



CGAGGGCAGCCAGCCAGCAGCUUCA







(SEQ ID NO: 1407)



GAGCUGAAAGAUGUCUACAACCAUU







(SEQ ID NO: 1408)



CACUAUGCCUAUUCCUUCAAGUAUU







(SEQ ID NO: 1409)



CCACUGACAUCAGCAUAGAUUUGAA







TAF1



(SEQ ID NO: 1410)



GAGGCACCUUUGGAGGGAAUAUUAU







(SEQ ID NO: 1411)



GAGGGAAUAUUAUCCAGCAUUCAAU







(SEQ ID NO: 1412)



GCAACCAAGAUAAAGAACUAUUAUA







(SEQ ID NO: 1413)



GAACAAGGUUCUGUCAUCAACUGAA







(SEQ ID NO: 1414)



CCACUGGACGCUGUCUCAAGAUUUA







TAF1D



(SEQ ID NO: 1415)



GGAUAAAUCAGGAAUAGAUUCUCUU







(SEQ ID NO: 1416)



CAUCUGAUGCUGUGGAACUUGCAAA







(SEQ ID NO: 1417)



AAAUCGAAGUGAUAACUCUUCUGAU







(SEQ ID NO: 1418)



GAUAACUCUUCUGAUAGCAGCUUAU







(SEQ ID NO: 1419)



ACUCUUCUGAUAGCAGCUUAUUUAA







TBC1D5



(SEQ ID NO: 1420)



GAAGAACUAUUUGUAAACAACAAUU







(SEQ ID NO: 1421)



GAACUAUUUGUAAACAACAAUUACU







(SEQ ID NO: 1422)



GGAAGGUUGUUGGCCAACAAGAUUU







(SEQ ID NO: 1423)



CAUGCACGAACUGUUAGCACCUAUA







(SEQ ID NO: 1424)



CCUAUGCAGUGUUCUCACAACUUAU







TEX2



(SEQ ID NO: 1425)



CACCUGAUUCCAAACUGAACUUACA







(SEQ ID NO: 1426)



CGACGCCUUUCAGAAGUCAUCUAUG







(SEQ ID NO: 1427)



CAGCAUCAAGGAGGAGGAGUGUGAU







(SEQ ID NO: 1428)



CAAGGAGGAGGAGUGUGAUUCUGAG







(SEQ ID NO: 1429)



CCAGUGAAGACGUUGGGCUUCUUUA







TEX21



(SEQ ID NO: 1430)



CCUUCAAGACCUUAUUGACUCUCUA







(SEQ ID NO: 1431)



CCCUGUGAAGGUGAGGGACAAUAAU







(SEQ ID NO: 1432)



CAACAGGAUUUGGUCUACAAAGAAA







(SEQ ID NO: 1433)



AAACGAGCAGCUGUGGAAAUUUAAA







(SEQ ID NO: 1434)



CCACCUUUAUGAAGAGGCUGGUGAA







TMEM49



(SEQ ID NO: 1435)



UUGGUGCAACCCUAAUUGGAA







(SEQ ID NO: 1436)



CUGGUUGUCCUGGAUGUUUGA







(SEQ ID NO: 1437)



UAGGGUGGAAUGUGAUGUUCA







(SEQ ID NO: 1438)



GAGACGUGUAGCAAUGAACAA







TNPO1



(SEQ ID NO: 1439)



CAAGAUCAUUGAGUGGUCUUAUCUU







(SEQ ID NO: 1440)



CCCAAAUGGUGUAACAGACUUUAUU







(SEQ ID NO: 1441)



CCAAAUGGUGUAACAGACUUUAUUA







(SEQ ID NO: 1442)



CAGACAUAGAUAUUAUCCUACUUAA







(SEQ ID NO: 1443)



CAGAUUCAGUAGGACAUCAUUUAAA







TRAF7



(SEQ ID NO: 1444)



CACUCCAAGUACGGGUGCACGUUCA







(SEQ ID NO: 1445)



CGGAGAAGAUCGACCAGCUAGAGAA







(SEQ ID NO: 1446)



GAGAAGAGCCUGGAGCUCAAGUUUG







(SEQ ID NO: 1447)



GACCCUCAGCAGAUCUUCAAGUGCA







(SEQ ID NO: 1448)



CCUGUGUGGUGUCUCUGCGUCUACU







TRIM66



(SEQ ID NO: 1449)



GGUGGUCCCGGAGACUUCACCUUGU







(SEQ ID NO: 1450)



CAGCGAUUGCUGGAGACAAGUUGUA







(SEQ ID NO: 1451)



CAGCUAGCUUCUCUUGGCUGCAUAA







(SEQ ID NO: 1452)



CAGUCUCCAGCAGUGUGCUCCUCAU







(SEQ ID NO: 1453)



AGGAGCCAAUUAACCUCUCUGUGAA







TSGA13



(SEQ ID NO: 1454)



CGUGAGAAAGGAAUGGUUGUCAAUA







(SEQ ID NO: 1455)



GAGAAAGGAAUGGUUGUCAAUAGCA







(SEQ ID NO: 1456)



CAAUAGCAAAGAGAUUUCUGAUGCA







(SEQ ID NO: 1457)



CAGUCCAUCCAAAUUUGGCCCAGUA







(SEQ ID NO: 1458)



CAUCCAAAUUUGGCCCAGUACUAUA 







TUBD1



(SEQ ID NO: 1459)



CAACUGGGCAUAUGGUUACUCUGUU







(SEQ ID NO: 1500)



CAUGAAGAAUCUAUAAUGAACAUAA







(SEQ ID NO: 1501)



GAGCUUUCGUUACACAGAAUUUAGA







(SEQ ID NO: 1502)



CAGAAUUUAGAAGAUCAGUACUCAA







(SEQ ID NO: 1503)



CAGACGCCCUCCUUCUUCAUGAGAA







TYW3



(SEQ ID NO: 1504)



CAGGAAAUGGAAGGCGCAAUGUUUG







(SEQ ID NO: 1505)



UGGUAGAGCUUGUGCAGUUUCUGAA







(SEQ ID NO: 1506)



GCGCUGGCCGCAUCCUACUCCUUGA







(SEQ ID NO: 1507)



AAGAUGAUGUGAUUGUAGCUCUGAA







(SEQ ID NO: 1508)



UGCCACUUUGAAAUUUGAACCAUUU







U58



(SEQ ID NO: 1509)



UGAUGACUAUCUUAGGACACCUUUG







(SEQ ID NO: 1510)



UGACUAUCUUAGGACACCUUUGGAA







(SEQ ID NO: 1511)



GACUAUCUUAGGACACCUUUGGAAU







(SEQ ID NO: 1512)



ACUAUCUUAGGACACCUUUGGAAUA







(SEQ ID NO: 1513)



UCUUAGGACACCUUUGGAAUAACUA







UBA52



(SEQ ID NO: 1514)



CAGAUCUUUGUGAAGACCCUCACUG







(SEQ ID NO: 1515)



CCAGUGACACCAUUGAGAAUGUCAA







(SEQ ID NO: 1516)



CACCUGACCAGCAGCGUCUGAUAUU







(SEQ ID NO: 1517)



GCAGCGUCUGAUAUUUGCCGGCAAA







(SEQ ID NO: 1518)



CAGCUGGAGGAUGGCCGCACUCUCU







USP10



(SEQ ID NO: 1519)



GAUCUUCAGUUGAGCUUCCUCCAUA







(SEQ ID NO: 1520)



CGGCCACCUGGAUAUUACAGCUAUU







(SEQ ID NO: 1521)



CCACCUGGAUAUUACAGCUAUUUGA







(SEQ ID NO: 1522)



CAGUGCAGAGGAUGCAGAAUUUAUG







(SEQ ID NO: 1523)



CAGUGACAUUGUGCCUGACAGUCCU







VOF16



(SEQ ID NO: 1524)



GCAUCCCAUGUUCACUGCUAUGGAU







(SEQ ID NO: 1525)



GAUGCCUGUGUAUAUGGCUAAUACG







(SEQ ID NO: 1526)



GGCUAAUACGGAGGCUGCUGGAUCU







(SEQ ID NO: 1527)



UAAUACGGAGGCUGCUGGAUCUAUU







(SEQ ID NO: 1528)



GGAGGCUGCUGGAUCUAUUCCUUGA







WDR51B



(SEQ ID NO: 1529)



CAACGGCAAGCAACUUGCUACUGCU







(SEQ ID NO: 1530)



CCGUGAGACUCUGGAUUCCUGAUAA







(SEQ ID NO: 1531)



CAGAUUCCGUUGGAUUUGCAAAUUU







(SEQ ID NO: 1532)



CAGCAGGUUCUGAUCAAACUGUGAA







(SEQ ID NO: 1533)



CAGCUUCUUCAGAUGGUACCCUUAA







WDR81



(SEQ ID NO: 1534)



CAGGUGACAGCAGCCAGGACUUGAA







(SEQ ID NO: 1535)



CAGCUGCCACAGGUGGUCUUCUCUG







(SEQ ID NO: 1536)



CCGCCCUGCUGGACGAGCUGCAGAA







(SEQ ID NO: 1537)



CGUGAGCCAGCAGGAUGCCCACUUU







(SEQ ID NO: 1538)



CCGUGCGCCUCUGGCCGCUGUACAA







WDR82



(SEQ ID NO: 1539)



UCGGACAAGAUUAACUGCUUCGAUU







(SEQ ID NO: 1540)



CGGACAAGAUUAACUGCUUCGAUUU







(SEQ ID NO: 1541)



CAGAUACACUCAUGCAGCAAACACA







(SEQ ID NO: 1542)



GCAAACACAGUUGUUUACAGCUCUA







(SEQ ID NO: 1543)



CAUUCGUCUGAUUGAUGCAUUCAAA







WIPF2



(SEQ ID NO: 1544)



CACCUCCCACAUUUCAUCAGGCAAA







(SEQ ID NO: 1545)



GGCGCCCUCUUACAGGACAUUUGCA







(SEQ ID NO: 1546)



CAAGCUGAAGAAGGUGACCAACAUU







(SEQ ID NO: 1547)



GCUGAAGAAGGUGACCAACAUUAAU







(SEQ ID NO: 1548)



CAGCCCAAGGGAGGUCUCUUCCAAG







ZBTB37



(SEQ ID NO: 1549)



CACAUGUCCUUGAAUGAGAUGAGUA







(SEQ ID NO: 1550)



GAGAUGAGUACAGUCUCCAUUUCAG







(SEQ ID NO: 1551)



CACAGAUCCUGGAGGGCAUUCAUUU







(SEQ ID NO: 1552)



CAGAGUCUCACAGGGUUACACCAAA







(SEQ ID NO: 1553)



GCCUGAGAAUCAGCCUUCUGGAGAA







ZC3H4



(SEQ ID NO: 1554)



GAGCCAAUGGGAGACGACGACUAUG







(SEQ ID NO: 1555)



GCCAUGACAUCGAACUCCCAAAGAA







(SEQ ID NO: 1556)



GCCAGAGCUGAGAACUGCCCUUAUA







(SEQ ID NO: 1557)



CCAGAGCUGAGAACUGCCCUUAUAU







(SEQ ID NO: 1558)



CACACCACUGGGAACUGCAUCAAUG







ZHX2



(SEQ ID NO: 1559)



CAAGGUGGUUAUGAGUGCAAAUACU







(SEQ ID NO: 1560)



GAGGCCAACUUCAAGCUGAAGUUAA







(SEQ ID NO: 1561)



AGGCCAACUUCAAGCUGAAGUUAAU







(SEQ ID NO: 1562)



CAACUUCAAGCUGAAGUUAAUUAAA







(SEQ ID NO: 1563)



ACGCAAUAAUCAAACUGUCUUGGAA







ZMYND8



(SEQ ID NO: 1564)



CAGGAGGUGGUAGAGGGCAUGGAUA







(SEQ ID NO: 1565)



CCUGGCUUACUGAACAGUAACAAUA







(SEQ ID NO: 1566)



CGGAAUGAUUUCUACUGCUGGGUUU







(SEQ ID NO: 1567)



GAGGAGUCCAUGGACUUCCUGGAUA







(SEQ ID NO: 1568)



CAAGACGGGACAAGCAGGGAGUUUA







ZNF 143



(SEQ ID NO: 1569)



GAAGGCAUUUCGAUGUGAAUAUGAU







(SEQ ID NO: 1570)



CGGUCGGUCCUUUACAACAUCAAAU







(SEQ ID NO: 1571)



CCAGUUUGUACAAACAUCAUGUUGU







(SEQ ID NO: 1572)



CGAGGAGGAGCAGGAAGCCUUCUUU







(SEQ ID NO: 1573)



CAGGUCAAGGUGAAGAUGUUCUUAA







ZNF662



(SEQ ID NO: 1574)



CAGGGAUGUGAUGCUGGAGAAUUAU







(SEQ ID NO: 1575)



GCAGGUUAGGGAGAUGGCCUGGUUA







(SEQ ID NO: 1576)



GGGACGUAUGGAAAGUUCUACAAAU







(SEQ ID NO: 1577)



CAAGAAUGUGCUAAGGCCUUUGUUU







(SEQ ID NO: 1578)



GCUAAGGCCUUUGUUUGGAAGUCAA







ZWILCH



(SEQ ID NO: 1579)



GCAAGGCAGUUAAUUGGACUUUACA







(SEQ ID NO: 1580)



CAGGAGAGCCCAGAGGUCCUUUGAA







(SEQ ID NO: 1581)



CAGAGAACUGAAAUUUCUUCUUGUU







(SEQ ID NO: 1582)



CAGGAAUUUCUGAAUGACUUAAAUA







(SEQ ID NO: 1583)



GAGCAGUAGUGUGAUUUCAUACCAA






Antisense Nucleic Acids

Generally, the term “antisense” refers to a nucleic acid molecule capable of hybridizing to a portion of an RNA sequence (such as mRNA) by virtue of some sequence complementarity. The antisense nucleic acids disclosed herein can be oligonucleotides that are double-stranded or single-stranded, RNA or DNA or a modification or derivative thereof, which can be directly administered to a cell (for example by administering the antisense molecule to the subject), or which can be produced intracellularly by transcription of exogenous, introduced sequences (for example by administering to the subject a vector that includes the antisense molecule under control of a promoter).


Antisense nucleic acids are polynucleotides, for example nucleic acid molecules that are at least 6 nucleotides in length, at least 10 nucleotides, at least 15 nucleotides, at least 20 nucleotides, at least 100 nucleotides, at least 200 nucleotides, such as 6 to 100 nucleotides. However, antisense molecules can be much longer. In particular examples, the nucleotide is modified at one or more base moiety, sugar moiety, or phosphate backbone (or combinations thereof), and can include other appending groups such as peptides, or agents facilitating transport across the cell membrane (Letsinger et al., Proc. Natl. Acad. Sci. USA 1989, 86:6553-6; Lemaitre et al., Proc. Natl. Acad. Sci. USA 1987, 84:648-52; WO 88/09810) or blood-brain barrier (WO 89/10134), hybridization triggered cleavage agents (Krol et al., BioTechniques 1988, 6:958-76) or intercalating agents (Zon, Pharm. Res. 5:539-49, 1988). Additional modifications include those set forth in U.S. Pat. Nos. 7,176,296; 7,329,648; 7,262,489, 7,115,579; and 7,105,495.


Examples of modified base moieties include, but are not limited to: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, acetylcytosine, 5-(carboxyhydroxylmethyl)uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N˜6-sopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-S-oxyacetic acid, 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl)uracil, and 2,6-diaminopurine.


Examples of modified sugar moieties include, but are not limited to: arabinose, 2-fluoroarabinose, xylose, and hexose, or a modified component of the phosphate backbone, such as phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, or a formacetal or analog thereof.


In a particular example, an antisense molecule is an α-anomeric oligonucleotide. An a-anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other (Gautier et al., Nucl. Acids Res. 15:6625-41, 1987). The oligonucleotide can be conjugated to another molecule, such as a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent. Oligonucleotides can include a targeting moiety that enhances uptake of the molecule by host cells. The targeting moiety can be a specific binding molecule, such as an antibody or fragment thereof that recognizes a molecule present on the surface of the host cell.


In a specific example, antisense molecules that recognize a nucleic acid set forth herein, include a catalytic RNA or a ribozyme (for example see WO 90/11364; WO 95/06764; and Sarver et al., Science 247:1222-5, 1990). Conjugates of antisense with a metal complex, such as terpyridylCu (II), capable of mediating mRNA hydrolysis, are described in Bashkin et al. (Appl. Biochem Biotechnol. 54:43-56, 1995). In one example, the antisense nucleotide is a 2′-O-methylribonucleotide (Inoue et al., Nucl. Acids Res. 15:6131-48, 1987), or a chimeric RNA-DNA analogue (Inoue et al., FEBS Lett. 215:327-30, 1987).


Antisense molecules can be generated by utilizing the Antisense Design algorithm of Integrated DNA Technologies, Inc. (1710 Commercial Park, Coralville, Iowa 52241 USA; http://www.idtdna.com/Scitools/Applications/AntiSense/Antisense.aspx/)


Examples of antisense nucleic acid molecules that can be utilized to decrease expression in the methods of the present invention, include, but are not limited to:


Also provided are sequences comprising the antisense sequences set forth above that are not the full length mRNA for any of the genes listed in Table 1 and can be used as antisense sequences. Further provided are antisense sequences that overlap with the sequences set forth above and comprise a fragment of the above-mentioned sequences. As mentioned above, these antisense sequences are merely exemplary, as it is known to those of skill in the art that once a mRNA sequence is provided for example the mRNA sequences set forth in Table 1, it is routine to walk along the mRNA sequence to generate antisense sequences that decrease expression. Therefore, the methods of the present invention can utilize any antisense sequence that decreases the expression of a gene set forth in Table 1.


Antisense molecules can be generated by utilizing the Antisense Design algorithm of Integrated DNA Technologies, Inc. (1710 Commercial Park, Coralville, Iowa 52241 USA; http://www.idtdna.com/Scitools/Applications/AntiSense/Antisense.aspx/)


Examples of antisense nucleic acid molecules that can be utilized to decrease expression in the methods of the present invention, include, but are not limited to:

















MTAP



GCCACCGTACCAGAGTTTCCT



(SEQ ID NO: 1584)



GCCACCGTACCAGAGTTTCC



(SEQ ID NO: SEQ ID NO: 1585)



GTTCACTGTCCTCCACTCCT



(SEQ ID NO: 1586)



GTTCACTGTCCTCCACTCCTG



(SEQ ID NO: 1587)



GTCACCTTAGTCACTCCCTCT



(SEQ ID NO: 1588)







AHR



ACCTCGTGATCCACCCTCCT



(SEQ ID NO: 1589)



GCTCTGTTCCTTCCTCATCT



(SEQ ID NO: 1590)



GCTCTGTTCCTTCCTCATCTG



(SEQ ID NO: 1591)



TGCTCTGTTCCTTCCTCATCT



(SEQ ID NO: 1592)



CTCTGTTCCTTCCTCATCTGT



(SEQ ID NO: 1593)







AK5



GTCCACATTTCCTCTTTCCC



(SEQ ID NO: 1594)



CGTCCACATTTCCTCTTTCCC



(SEQ ID NO: 1595)



CCTCCCATAGTGTCCTCTCC



(SEQ ID NO: 1596)



TCCACATTTCCTCTTTCCC



(SEQ ID NO: 1597)



CCTCCCATAGTGTCCTCTCCA



(SEQ ID NO: 1598)







AMOTL2



GGGCTTCCTGGGTCTTGCTT



(SEQ ID NO: 1599)



ACCGCTCATTGTCCCTCTGC



(SEQ ID NO: 1600)



GCTTCCTGGGTCTTGCTTGC



(SEQ ID NO: 1601)



CCGCTCATTGTCCCTCTGCA



(SEQ ID NO: 1602)



ACCGCTCATTGTCCCTCTGCA



(SEQ ID NO: 1603)







ANKMY2



GCCCTTACTCCTCAGACACC



(SEQ ID NO: 1604)



GCCCTTACTCCTCAGACACCT



(SEQ ID NO: 1605)



TCTGCTTGCCTCTTTGTTCCC



(SEQ ID NO: 1606)



TCTCCTCTTCACTCTTTGCC



(SEQ ID NO: 1607)



CTGCTTGCCTCTTTGTTCCC



(SEQ ID NO: 1608)







ANXA4



TTCCTTCATCCCTCCCACCA



(SEQ ID NO: 1609)



TCCTTCATCCCTCCCACCA



(SEQ ID NO: 1610)



TTTCCTTCATCCCTCCCACC 



(SEQ ID NO: 1611)



TTTCCTTCATCCCTCCCACCA 



(SEQ ID NO: 1612)



TTCCTTCATCCCTCCCACC 



(SEQ ID NO: 1613)







ARL6IP5 



GCTCGCCAACATGACCACCA



(SEQ ID NO: 1614)



TTATTGTCCTCCTTGTGCCC



(SEQ ID NO: 1615)



CCACTCCAGTTGTCTTCACA



(SEQ ID NO: 1616)



GCCCATCGGTGTCCTCTTCA



(SEQ ID NO: 1617)



ATTATTGTCCTCCTTGTGCCC



(SEQ ID NO: 1618)







ARSA



GCCACTTGCCAGCTATCCCT



(SEQ ID NO: 1619)



CCACTTGCCAGCTATCCCT



(SEQ ID NO: 1620)



CCACTTGCCAGCTATCCCTG



(SEQ ID NO: 1621)



GTGTGCCTTGTACTTCCCGCT



(SEQ ID NO: 1622)



GTGCCTTGTACTTCCCGCTC



(SEQ ID NO: 1623)







ATOH8



GTCTCCAAGTGTCTCTGCCA



(SEQ ID NO: 1624)



CGTCTCCAAGTGTCTCTGCCA



(SEQ ID NO: 1625)



GTCTCCAAGTGTCTCTGCCAT



(SEQ ID NO: 1626)



CTGCTCCACACACTCTCTGA



(SEQ ID NO: 1627)



CTGCTCCACACACTCTCTG



(SEQ ID NO: 1628)







ATP6V1A



CATTTCAGCCAGTCGTCCC



(SEQ ID NO: 1629)



GCATTTCAGCCAGTCGTCCC



(SEQ ID NO: 1630)



CCCACTCACACATCACTCACT



(SEQ ID NO: 1631)



CATTTCAGCCAGTCGTCCCG



(SEQ ID NO: 1632)



TTTCCTCGTTCTCCACAGCC



(SEQ ID NO: 1633)







BPNT1



TTTCCCATTTCTCCACCT  



(SEQ ID NO: 1634)



TTTCCTGCTCCTCCTACTCG



(SEQ ID NO: 1635)



ATTTCCTGCTCCTCCTACTC



(SEQ ID NO: 1636)



TTCCTGCTCCTCCTACTCG



(SEQ ID NO: 1637)



TTTCCTGCTCCTCCTACTC



(SEQ ID NO: 1638)







C11ORF54



GCTCCCTCTGACCACCTATTC



(SEQ ID NO: 1639)



CTCCCTCTGACCACCTATTCC



(SEQ ID NO: 1640)



GCTCCCTCTGACCACCTATT



(SEQ ID NO: 1641)



GCTCCCTCTGACCACCTAT



(SEQ ID NO: 1642)



TCCCTCTGACCACCTATTCCA



(SEQ ID NO: 1643)







C17ORF75



GCCCAGCCACTCTCACTTT



(SEQ ID NO: 1644)



GTGCCCAGCCACTCTCACTT



(SEQ ID NO: 1645)



GCCCAGCCACTCTCACTTTA



(SEQ ID NO: 1646)



GTGCCCAGCCACTCTCACTTT



(SEQ ID NO: 1647)



TGCCCAGCCACTCTCACTTT



(SEQ ID NO: 1648)







C18ORF32



CCCTTACAGTCCACTTTGCCT



(SEQ ID NO: 1649)



CCCTTACAGTCCACTTTGCC



(SEQ ID NO: 1650)



ACCCTTACAGTCCACTTTGCC



(SEQ ID NO: 1651)



GTCCACTTTGCCTTTGTTTG



(SEQ ID NO: 1652)



GTCCACTTTGCCTTTGTTT



(SEQ ID NO: 1653)







C1ORF116



CCTACTCTTACCTCCCTCCC



(SEQ ID NO: 1654)



CCCTACTCTTACCTCCCTCC



(SEQ ID NO: 1655)



ACCCTACTCTTACCTCCCTCC



(SEQ ID NO: 1656)



ACCCTACTCTTACCTCCCTC



(SEQ ID NO: 1657)



CTACTCTTACCTCCCTCCC



(SEQ ID NO: 1658)







C6ORF176



GTTCCTCCTTCCCTCCTTTCC



(SEQ ID NO: 1659)



TCCTCCTTCCCTCCTTTCCA



(SEQ ID NO: 1660)



GTTCCTCCTTCCCTCCTTTC



(SEQ ID NO: 1661)



TTCCTCCTTCCCTCCTTTCC



(SEQ ID NO: 1662)



TCCTCCTTCCCTCCTTTCC



(SEQ ID NO: 1663)







C6ORF62



GCCAATTACATCCCTTCTCA



(SEQ ID NO: 1664)



GCCAATTACATCCCTTCTCAT



(SEQ ID NO: 1665)



GCCAATTACATCCCTTCTC



(SEQ ID NO: 1666)



CAGCCAATTACATCCCTTCTC



(SEQ ID NO: 1667)



AGCCAATTACATCCCTTCTCA



(SEQ ID NO: 1668)







CBLN2



CCAAGTCCTCCCAGCCACAT



(SEQ ID NO: 1669)



CAAGTCCTCCCAGCCACAT



(SEQ ID NO: 1670)



CAAGTCCTCCCAGCCACATG



(SEQ ID NO: 1671)



AAGTCCTCCCAGCCACATG



(SEQ ID NO: 1672)



GTCTGTGTGTCTGCTCCTCT



(SEQ ID NO: 1673)







CCDC57



TCCACTCCTCCTCCTTGCGA



(SEQ ID NO: 1674)



TCCACTCCTCCTCCTTGCGAA



(SEQ ID NO: 1675)



GTTTCCTCTTCCTTCTTCCT



(SEQ ID NO: 1676)



CACCACCTCCTGCTCCTGTA



(SEQ ID NO: 1677)



CCACTCCTCCTCCTTGCGAA



(SEQ ID NO: 1678)







CDH9



CTGTCTTCTCTTGCCTCTGGA



(SEQ ID NO: 1679)



CTGTCTTCTCTTGCCTCTGG



(SEQ ID NO: 1680)



CTGTCTTCTCTTGCCTCTG



(SEQ ID NO: 1681)



TTCCCACAGAGGCACAGTCC



(SEQ ID NO: 1682)



TTTCCCACAGAGGCACAGTC



(SEQ ID NO: 1683)







CEP152



CCTCATGCTGTTCTTCCCACT



(SEQ ID NO: 1684)



CCACCTTCATCTCCACCTTCT



(SEQ ID NO: 1685)



CTTCCACCTTCATCTCCACCT



(SEQ ID NO: 1686)



TCCACCTTCATCTCCACCTTC



(SEQ ID NO: 1687)



CCACTTCCACCTTCATCTCCA



(SEQ ID NO: 1688)







CKAP2L



GCCGTGACTTGTTCCCTCTCT



(SEQ ID NO: 1689)



GCCGTGACTTGTTCCCTCTC



(SEQ ID NO: 1690)



TCGCCGTGACTTGTTCCCTCT



(SEQ ID NO: 1691)



CGCCGTGACTTGTTCCCTCT



(SEQ ID NO: 1692)



GCCGTGACTTGTTCCCTCT



(SEQ ID NO: 1693)







CLTC



CATTCATTCGTCCTCCCTTCT



(SEQ ID NO: 1694)



TCATTCATTCGTCCTCCCTTC



(SEQ ID NO: 1695)



TTCATTCGTCCTCCCTTCTT



(SEQ ID NO: 1696)



TCATTCGTCCTCCCTTCTTG



(SEQ ID NO: 1697)



TTCATTCGTCCTCCCTTCTTG



(SEQ ID NO: 1698)







CLUL1



GCCCTCTCATCTTCTCCACC



(SEQ ID NO: 1699)



GCCCTCTCATCTTCTCCACCA



(SEQ ID NO: 1700)



CCCTCTCATCTTCTCCACCA



(SEQ ID NO: 1701)



CCCTCTCATCTTCTCCACC



(SEQ ID NO: 1702)



CCCTCTCATCTTCTCCACCAG



(SEQ ID NO: 1703)







CNTN5



TCACATGCCACTCCTCTCCA  



(SEQ ID NO: 1704)



ATCACATGCCACTCCTCTCCA



(SEQ ID NO: 1705)



ATCACATGCCACTCCTCTCC  



(SEQ ID NO: 1706)



TCACATGCCACTCCTCTCC



(SEQ ID NO: 1707)



CACATGCCACTCCTCTCCA



(SEQ ID NO: 1708)







CRYZ



TCCAACTCCTCCACTTGCCC  



(SEQ ID NO: 1709)



ATCCAACTCCTCCACTTGCCC 



(SEQ ID NO: 1710)



ATCCAACTCCTCCACTTGCC 



(SEQ ID NO: 1711)



TCCAACTCCTCCACTTGCC



(SEQ ID NO: 1712)



CTGCCACACCTTCTCCAACT 



(SEQ ID NO: 1713)







DAB2



GCATCACCCTCTTCTTCCC



(SEQ ID NO: 1714)



TCCTTTGTTTGTGTTGTCCCT



(SEQ ID NO: 1715)



CCTTTGTTTGTGTTGTCCCT  



(SEQ ID NO: 1716)



AGCATCACCCTCTTCTTCCC 



(SEQ ID NO: 1717)



TCCTTTGTTTGTGTTGTCCC  



(SEQ ID NO: 1718)







DARS2



CCCTTCTCCTCTTTCCACGCT



(SEQ ID NO: 1719)



CCCTTCTCCTCTTTCCACGC  



(SEQ ID NO: 1720)



GCCCTTCTCCTCTTTCCACG 



(SEQ ID NO: 1721)



CGCCCTTCTCCTCTTTCCAC 



(SEQ ID NO: 1722)



GCCCTTCTCCTCTTTCCAC



(SEQ ID NO: 1723)







DDIT4



TCCTGCCTCTAGTCTCCACC  



(SEQ ID NO: 1724)



CTCCTGCCTCTAGTCTCCAC 



(SEQ ID NO: 1725)



TCCTGCCTCTAGTCTCCAC



(SEQ ID NO: 1726)



CTCCTGCCTCTAGTCTCCA



(SEQ ID NO: 1727)



TGCCTCTTCCTTCACCCTGG  



(SEQ ID NO: 1728)







DDX42



TCCCACATACTCCCTCCTCC



(SEQ ID NO: 1729)



TCCCACATACTCCCTCCTCCA



(SEQ ID NO: 1730)



CCCACATACTCCCTCCTCCA



(SEQ ID NO: 1731)



CTCCCACATACTCCCTCCTC



(SEQ ID NO: 1732)



GCTCCCACATACTCCCTCCT



(SEQ ID NO: 1733)







DNAH2



CTTCACTCCCTTCTTCACCA



(SEQ ID NO: 1734)



GCCCACTCCTTCACCCATTT



(SEQ ID NO: 1735)



TCCAGTTCTCCTCCCAGTCTC



(SEQ ID NO: 1736)



GCCCACTCCTTCACCCATT



(SEQ ID NO: 1737)



CAGCCCACTCCTTCACCCA



(SEQ ID NO: 1738)







DUSP5



GCCGATTCCTCCATCCCAGT



(SEQ ID NO: 1739)



GCCGATTCCTCCATCCCAGTT



(SEQ ID NO: 1740)



GCCTTCCTTCAGTCCCGAGA



(SEQ ID NO: 1741)



GGGCTCTCTCACTCTCAATCT



(SEQ ID NO: 1742)



TCTTTCTCCCACGTTGCTGC



(SEQ ID NO: 1743)







EBNA1BP2



GCTTTCTCCTGCTGCCTCTTC



(SEQ ID NO: 1744)



ACCCATTCCAGATCCCGCTT



(SEQ ID NO: 1745)



ACCCATTCCAGATCCCGCT



(SEQ ID NO: 1746)



CCTTCTTCCCGTATTTCCT



(SEQ ID NO: 1747)



CCCATTCCAGATCCCGCTT



(SEQ ID NO: 1748)







EEF1A1



TTCCCATCTCAGCAGCCTCC



(SEQ ID NO: 1749)



CCCTTTCCCATCTCAGCAG



(SEQ ID NO: 1750)



CCTTTCCCATCTCAGCAGCCT



(SEQ ID NO: 1751)



TTTCCCATCTCAGCAGCCTCC



(SEQ ID NO: 1752)



CCTTTCCCATCTCAGCAGCC



(SEQ ID NO: 1753)







EID3



CTCCTTTCTCTTCGTCTCCCT



(SEQ ID NO: 1754)



TCTCCTTTCTCTTCGTCTCCC



(SEQ ID NO: 1755)



CTCCTTTCTCTTCGTCTCCC



(SEQ ID NO: 1756)



TCCTTTCTCTTCGTCTCCCT



(SEQ ID NO: 1757)



CTTTCTCTTCGTCTCCCTTCC



(SEQ ID NO: 1758)







ENO1



GCCTTTGAGACACCCTTCCC



(SEQ ID NO: 1759)



AGCCTTTGAGACACCCTTCCC



(SEQ ID NO: 1760)



CCTTTGAGACACCCTTCCC



(SEQ ID NO: 1761)



GCATCTTTCCCATATTTCTCC



(SEQ ID NO: 1762)



GTCCATGCCGATGACCACCT



(SEQ ID NO: 1763)







ERGIC1



CCCACTTCCCACACACCCTT



(SEQ ID NO: 1764)



CCCACTTCCCACACACCCTTT



(SEQ ID NO: 1765)



CCACTTCCCACACACCCTTT



(SEQ ID NO: 1766)



CCACTTCCCACACACCCTT



(SEQ ID NO: 1767)



CTGCTTGCCACTCTTGTCCTC



(SEQ ID NO: 1768)







FAM126B



CAGCAGTCTACCACCTTTCCC



(SEQ ID NO: 1769)



AGCAGTCTACCACCTTTCCC



(SEQ ID NO: 1770)



GCAGTCTACCACCTTTCCC



(SEQ ID NO: 1771)



AGTACCTCCCTCAGTGCCA



(SEQ ID NO: 1772)



TAGTACCTCCCTCAGTGCCA



(SEQ ID NO: 1773)







FAM35A



TCTGCCACGACCATCCCAT



(SEQ ID NO: 1774)



TCTGCCACGACCATCCCATAG



(SEQ ID NO: 1775)



TCTGCCACGACCATCCCATA



(SEQ ID NO: 1776)



CTGCCACGACCATCCCATAG



(SEQ ID NO: 1777)



CTGCCACGACCATCCCATA



(SEQ ID NO: 1778)







FAM55C



TTCTCCCACCTCAGCCTCCT  



(SEQ ID NO: 1779)



ATTCTCCCACCTCAGCCTCCT 



(SEQ ID NO: 1780)



ATTCTCCCACCTCAGCCTCC 



(SEQ ID NO: 1781)



GTTCTTCCCTCCCACAATGCC



(SEQ ID NO: 1782)



CCACACCCTGCCATGACTCT 



(SEQ ID NO: 1783)







FKBP2



TTCTCCCACCTCAGCCTCCT 



(SEQ ID NO: 1784)



ATTCTCCCACCTCAGCCTCCT 



(SEQ ID NO: 1785)



ATTCTCCCACCTCAGCCTCC  



(SEQ ID NO: 1786)



GTTCTTCCCTCCCACAATGCC



(SEQ ID NO: 1787)



CCACACCCTGCCATGACTCT 



(SEQ ID NO: 1788)







FNDC3A



ACTTCCTTCAACTCCTCCC



(SEQ ID NO: 1789)



TACTTCCTTCAACTCCTCCC 



(SEQ ID NO: 1790)



ATACTTCCTTCAACTCCTCC  



(SEQ ID NO: 1791)



CTCCCGCTTTCTGTTTCACCC



(SEQ ID NO: 1792)



CCTTCTGCCATCTCCACTACA



(SEQ ID NO: 1793)







FNDC3B



ACTTCCTTCAACTCCTCCC



(SEQ ID NO: 1794)



TACTTCCTTCAACTCCTCCC 



(SEQ ID NO: 1795)



ATACTTCCTTCAACTCCTCCC 



(SEQ ID NO: 1796)



CTCCCGCTTTCTGTTTCACCC



(SEQ ID NO: 1797)



CCTTCTGCCATCTCCACTACA



(SEQ ID NO: 1798)







G2E3



TCCCACCTCAGCCTCCCTTA 



(SEQ ID NO: 1799)



ATTCTCCCACCTCAGCCTCC 



(SEQ ID NO: 1800)



TCCCACCTCAGCCTCCCTTAT 



(SEQ ID NO: 1801)



CCCACCTCAGCCTCCCTTAT  



(SEQ ID NO: 1802)



CCCACCTCAGCCTCCCTTATA



(SEQ ID NO: 1803)







GDF15



ACCGTCCTGAGTTCTTGCCC



(SEQ ID NO: 1804)



CAGTTCCATCAGACCAGCCC



(SEQ ID NO: 1805)



GCCATTCACCGTCCTGAGTTC



(SEQ ID NO: 1806)



GAGCCATTCACCGTCCTGAGT



(SEQ ID NO: 1807)



AGTTCCATCAGACCAGCCC



(SEQ ID NO: 1808)







GEN1



CCTTCCCTCTGTCTCTGCT



(SEQ ID NO: 1809)



CCTTCCCTCTGTCTCTGCTA



(SEQ ID NO: 1810)



ACCTTCCCTCTGTCTCTGCT



(SEQ ID NO: 1811)



CCTTCCCTCTGTCTCTGCTAT



(SEQ ID NO: 1812)



ACCTTCCCTCTGTCTCTGCTA



(SEQ ID NO: 1813)







GLIS2



GTCCTCTCCCGCTTCTCTCT



(SEQ ID NO: 1814)



GTCCTCTCCCGCTTCTCTCTT



(SEQ ID NO: 1815)



TCCTCTCCCGCTTCTCTCTT



(SEQ ID NO: 1816)



TCCTCTCCCGCTTCTCTCT



(SEQ ID NO: 1817)



TCCTCTCCCGCTTCTCTCTTG



(SEQ ID NO: 1818)







GLUD1



CTGCTCTTGACTGTTCCTCCC



(SEQ ID NO: 1819)



GCTCTTGACTGTTCCTCCC



(SEQ ID NO: 1820)



TGCTCTTGACTGTTCCTCCC



(SEQ ID NO: 1821)



GTCACTCCTCCAGCATTCA



(SEQ ID NO: 1822)



GTAGCCTTCGATGACCTCCCA



(SEQ ID NO: 1823)







GLYCTK



CCACCTTCTCAACTCTGCTCC



(SEQ ID NO: 1824)



GCCACCTTCTCAACTCTGCTC



(SEQ ID NO: 1825)



CCACCTTCTCAACTCTGCTC



(SEQ ID NO: 1826)



GCCACCTTCTCAACTCTGCT



(SEQ ID NO: 1827)



CCACCTTCTCAACTCTGCT



(SEQ ID NO: 1828)







GTF2H5



AATCCTCCCACCTCAGCCTC



(SEQ ID NO: 1829)



CAATCCTCCCACCTCAGCCT



(SEQ ID NO: 1830)



AATCCTCCCACCTCAGCCT



(SEQ ID NO: 1831)



TCCCACCTCAGCCTCCCTAA



(SEQ ID NO: 1832)



AGCAATCCTCCCACCTCAGC



(SEQ ID NO: 1833)







HAVCR1



ACCTGCCTCTCCACCAACCT



(SEQ ID NO: 1834)



GTTCTCTCCTTATTGCTCCCT



(SEQ ID NO: 1835)



CCTGCCTCTCCACCAACCTT



(SEQ ID NO: 1836)



ACCTGCCTCTCCACCAACCTT



(SEQ ID NO: 1837)



CCTGCCTCTCCACCAACCTTT



(SEQ ID NO: 1838)







HLA-DMA



GACCCATACCTTCTTGCCACA



(SEQ ID NO: 1839)



CCCATACCTTCTTGCCACACA



(SEQ ID NO: 1840)



GACCCATACCTTCTTGCCAC



(SEQ ID NO: 1841)



CCCATACCTTCTTGCCACAC



(SEQ ID NO: 1842)



ACCCATACCTTCTTGCCACAC



(SEQ ID NO: 1843)







HNRNPH3



ACATCCCACGCCATCCACCT



(SEQ ID NO: 1844)



ACGCCATCCACCTCCACTCA



(SEQ ID NO: 1845)



TACATCCCACGCCATCCACCT



(SEQ ID NO: 1846)



CGCCATCCACCTCCACTCAT



(SEQ ID NO: 1847)



GTCCTCCCATACCTCTTCCA



(SEQ ID NO: 1848)







HNRNPK



GCCCTCCTCCTTTCTCGTTC



(SEQ ID NO: 1849)



GCCCTCCTCCTTTCTCGTT



(SEQ ID NO: 1850)



TCCACTCACTCTGCTGCTGT



(SEQ ID NO: 1851)



TCCACTCACTCTGCTGCTGTT



(SEQ ID NO: 1852)



CCCTCCTCCTTTCTCGTTC



(SEQ ID NO: 1853)







HSP90AB4P



CCCTTCTTCCACTCTTGCTCC



(SEQ ID NO: 1854)



CCTTCTTCCACTCTTGCTCC  



(SEQ ID NO: 1855)



CCCTTCTTCCACTCTTGCTC 



(SEQ ID NO: 1856)



CCTTCTTCCACTCTTGCTCCA



(SEQ ID NO: 1857)



GCCCTTCTTCCACTCTTGCTC



(SEQ ID NO: 1858)







IARS2



CCCTTCTTCCACTCTTGCTCC 



(SEQ ID NO: 1859)



CCTTCTTCCACTCTTGCTCC  



(SEQ ID NO: 1860)



CCCTTCTTCCACTCTTGCTC 



(SEQ ID NO: 1861)



CCTTCTTCCACTCTTGCTCCA 



(SEQ ID NO: 1862)



GCCCTTCTTCCACTCTTGCTC 



(SEQ ID NO: 1863)







IL18



CTCACCACAACCTCTACCTCC



(SEQ ID NO: 1864)



TCACCACAACCTCTACCTCC 



(SEQ ID NO: 1865)



GTTCCTTTCCTCTTCCCGA



(SEQ ID NO: 1866)



CTCACCACAACCTCTACCTC  



(SEQ ID NO: 1867)



CACCACAACCTCTACCTCC



(SEQ ID NO: 1868)







IL6ST



TCCCTTCCACCATCCCACTC 



(SEQ ID NO: 1869)



TCCCTTCCACCATCCCACTCA 



(SEQ ID NO: 1870)



CCCTTCCACCATCCCACTCA  



(SEQ ID NO: 1871)



TTCCCTTCCACCATCCCACTC 



(SEQ ID NO: 1872)



TCCACCATCCCACTCACACCT



(SEQ ID NO: 1873)







ITGB1



GCCACCAAGTTTCCCATCTCC 



(SEQ ID NO: 1874)



GCCACCAAGTTTCCCATCTC 



(SEQ ID NO: 1875)



TGCCACCAAGTTTCCCATCTC 



(SEQ ID NO: 1876)



GCCACCAAGTTTCCCATCT



(SEQ ID NO: 1877)



TGCCACCAAGTTTCCCATCT  



(SEQ ID NO: 1878)







KDM6B



GCCACCAAGTTTCCCATCTCC



(SEQ ID NO: 1879)



GCCACCAAGTTTCCCATCTC



(SEQ ID NO: 1880)



TGCCACCAAGTTTCCCATCTC



(SEQ ID NO: 1881)



GCCACCAAGTTTCCCATCT



(SEQ ID NO: 1882)



TGCCACCAAGTTTCCCATCT



(SEQ ID NO: 1883)







KAZALD1



TCTCTCCCTACCCTCCTCCA



(SEQ ID NO: 1884)



TTCTCTCCCTACCCTCCTCC



(SEQ ID NO: 1885)



TTCTCTCCCTACCCTCCTCCA



(SEQ ID NO: 1886)



CTTCTCTCCCTACCCTCCTC



(SEQ ID NO: 1887)



GCTTCTCTCCCTACCCTCCT



(SEQ ID NO: 1888)







KCTD1



CCCAACTCCCTTTCCGACTCT



(SEQ ID NO: 1889)



GTCAGCCACATCCCTTTCG



(SEQ ID NO: 1890)



CGTCAGCCACATCCCTTTC



(SEQ ID NO: 1891)



ACTCCCTTTCCGACTCTTGC



(SEQ ID NO: 1892)



GTCAGCCACATCCCTTTCGA



(SEQ ID NO: 1893)







KIAA1199



ACTCTCCTCTCCATCACCACT



(SEQ ID NO: 1894)



ACTCTCCTCTCCATCACCAC



(SEQ ID NO: 1895)



CTCTCCTCTCCATCACCACT



(SEQ ID NO: 1896)



GTTCTCACCTCTCCATCCCG



(SEQ ID NO: 1897)



GTTCTCACCTCTCCATCCC



(SEQ ID NO: 1898)







KYNU



AGCCACCCTCTCATCCGTTG



(SEQ ID NO: 1899)



AGCCACCCTCTCATCCGTT



(SEQ ID NO: 1900)



GAGCCACCCTCTCATCCGTT



(SEQ ID NO: 1901)



GCCACTAGATGTCACCCTTT



(SEQ ID NO: 1902)



AGCCACTAGATGTCACCCTT



(SEQ ID NO: 1903)







LMX1B



TCTCCCTCTTCCCACTCCA



(SEQ ID NO: 1904)



ATCTCCCTCTTCCCACTCCA



(SEQ ID NO: 1905)



TCTCCCTCTTCCCACTCCAG



(SEQ ID NO: 1906)



ATCTCCCTCTTCCCACTCC



(SEQ ID NO: 1907)



TATCTCCCTCTTCCCACTCC



(SEQ ID NO: 1908)







LNX2



GCTCACCACTCTTACTTCCC



(SEQ ID NO: 1909)



CTCACCACTCTTACTTCCC



(SEQ ID NO: 1910)



AGCTCACCACTCTTACTTCCC



(SEQ ID NO: 1911)



GTCCCGTTTATGAAGAGCCAC



(SEQ ID NO: 1912)



GCTCACCACTCTTACTTCC



(SEQ ID NO: 1913)







MALAT1



CCACCCTCTCTCTTCCCTGT



(SEQ ID NO: 1914)



CCACCCTCTCTCTTCCCTGTT



(SEQ ID NO: 1915)



TGCTGTTACCTCCCACCTCC



(SEQ ID NO: 1916)



CCCTCTCTCTTCCCTGTTA



(SEQ ID NO: 1917)



TTTCATCCTACCACTCCCA



(SEQ ID NO: 1918)







MAPK1IP1L



GTCTTCCCTGAACGCCACCT



(SEQ ID NO: 1919)



AGTCTTCCCTGAACGCCACCT



(SEQ ID NO: 1920)



GTCTTCCCTGAACGCCACCTA



(SEQ ID NO: 1921)



AGTCTTCCCTGAACGCCACC



(SEQ ID NO: 1922)



TCTTCCCTGAACGCCACCTAC



(SEQ ID NO: 1923)







MAP4



TCCACCTCCACTCTTCCCTC



(SEQ ID NO: 1924)



CACCTCCACTCTTCCCTCCA



(SEQ ID NO: 1925)



ACCTCCACTCTTCCCTCCA



(SEQ ID NO: 1926)



TCCACCTCCACTCTTCCCT



(SEQ ID NO: 1927)



ATCCACCTCCACTCTTCCCTC



(SEQ ID NO: 1928)







MGAT3



CATCCCTCAGCACCTCCTCT 



(SEQ ID NO: 1929)



TCCCTCAGCACCTCCTCTTG 



(SEQ ID NO: 1930)



CATCCCTCAGCACCTCCTCTT



(SEQ ID NO: 1931)



TCCCTCAGCACCTCCTCTT



(SEQ ID NO: 1932)



ATCCCTCAGCACCTCCTCTT  



(SEQ ID NO: 1933)







MICALCL



CCCTCCCTCTTCTTCCTTCCT 



(SEQ ID NO: 1934)



CCCTCCCTCTTCTTCCTTCC 



(SEQ ID NO: 1935)



CCTCCCTCTTCTTCCTTCCT 



(SEQ ID NO: 1936)



CCTCCCTCTTCTTCCTTCCTG 



(SEQ ID NO: 1937)



CCTCCCTCTTCTTCCTTCC



(SEQ ID NO: 1938)







MIR194-1



CCACATGGAGTTGCTGTTACA



(SEQ ID NO: 1939)



CCACATGGAGTTGCTGTTAC 



(SEQ ID NO: 1940)



GGAGTTGCTGTTACACTTGA  



(SEQ ID NO: 1941)



GGAGTTGCTGTTACACTTGAT



(SEQ ID NO: 1942)



TGGAGTTGCTGTTACACTTGA



(SEQ ID NO: 1943)







MIR215



GTCTGTCAATTCATAGGTC



(SEQ ID NO: 1944)



GAAGTAGCACAGTCATACAG  



A (SEQ ID NO: 1945)



AGTAGCACAGTCATACAGA



(SEQ ID NO: 1946)



GCCTAAAGAAATGACAGAC



(SEQ ID NO: 1947)



ATGACAGACAAACTCAGCT



(SEQ ID NO: 1948)







MIR632



TACCACCACGTCCCACAGGA 



(SEQ ID NO: 1949)



ACCACGTCCCACAGGAAGCA 



(SEQ ID NO: 1950)



CCCACAGGAAGCAGACACA



(SEQ ID NO: 1951)



ACCACCACGTCCCACAGGAA 



(SEQ ID NO: 1952)



TACCACCACGTCCCACAGGA 



A (SEQ ID NO: 1953)







MIRLET7G



GTTATCTCCTGTACCGGGTGG



(SEQ ID NO: 1954)



GTACAAACTACTACCTCAGCC



(SEQ ID NO: 1955)



GTGGCCTGTACAGTTATCTCC



(SEQ ID NO: 1956)



GCCTGTACAGTTATCTCCTG



(SEQ ID NO: 1957)



ACAAACTACTACCTCAGCCT



(SEQ ID NO: 1958)







MIRN15A



TGTGCTGCTACTTTACTCCA



(SEQ ID NO: 1959)



GTGCTGCTACTTTACTCCA



(SEQ ID NO: 1960)



ATGTGCTGCTACTTTACTCCA



(SEQ ID NO: 1961)



ATGTGCTGCTACTTTACTCC



(SEQ ID NO: 1962)



TGTGCTGCTACTTTACTCC



(SEQ ID NO: 1963)







MIRN16-1



ACCTTACTTCAGCAGCACAGT



(SEQ ID NO: 1964)



CCTTACTTCAGCAGCACAGTT



(SEQ ID NO: 1965)



CCTTACTTCAGCAGCACAGT



(SEQ ID NO: 1966)



GTCAACCTTACTTCAGCAGCA



(SEQ ID NO: 1967)



GTCAACCTTACTTCAGCAG



(SEQ ID NO: 1968)







MIRN16-2



TTTACGTGCTGCTAGAGTGGA



(SEQ ID NO: 1969)



TTACGTGCTGCTAGAGTGGA



(SEQ ID NO: 1970)



TACGTGCTGCTAGAGTGGA



(SEQ ID NO: 1971)



CGTGCTGCTAGAGTGGAAC



(SEQ ID NO: 1972)



ACGTGCTGCTAGAGTGGAAC



(SEQ ID NO: 1973)







MLF1IP



CCCACTCTCTATCCCTTCACC



(SEQ ID NO: 1974)



TCCCACTCTCTATCCCTTCAC



(SEQ ID NO: 1975)



TCCCACTCTCTATCCCTTCA



(SEQ ID NO: 1976)



CCACTCTCTATCCCTTCACCA



(SEQ ID NO: 1977)



CCTATCCCACTCTCTATCCCT



(SEQ ID NO: 1978)







MLL5



TGCCTCCCTCTGAACACCCT



(SEQ ID NO: 1979)



TTCTTTGTGCCTCCCTCTGA



(SEQ ID NO: 1980)



CTATTCTTTGTGCCTCCCTCT



(SEQ ID NO: 1981)



TCTTTGTGCCTCCCTCTGA



(SEQ ID NO: 1982)



ATTCTTTGTGCCTCCCTCTG



(SEQ ID NO: 1983)







MRPL45



ACTCTGATCCCTGCCCTGTGA



(SEQ ID NO: 1984)



ACTCTGATCCCTGCCCTGTG



(SEQ ID NO: 1985)



ACTCTGATCCCTGCCCTGT



(SEQ ID NO: 1986)



CTCTGATCCCTGCCCTGTGA



(SEQ ID NO: 1987)



GTTCTCTCTATCAGTCCCTCC



(SEQ ID NO: 1988)







MRPS18B



ATCTGTACCACCCGCCCTCA



(SEQ ID NO: 1989)



GTCCCTCTAGCTCTCTTCCC



(SEQ ID NO: 1990)



GCCTCTCTTCCCAGTCTACA



(SEQ ID NO: 1991)



GTCCCTCTAGCTCTCTTCCCA



(SEQ ID NO: 1992)



CCTCTCTTCCCAGTCTACAGC



(SEQ ID NO: 1993)







MTHFD1



GCCCAGTCTCTCTCTCATGTC



(SEQ ID NO: 1994)



GCCCAGTCTCTCTCTCATGT



(SEQ ID NO: 1995)



TCCGTGTGTGACCCTTCTCC



(SEQ ID NO: 1996)



TGCCCAGTCTCTCTCTCATGT



(SEQ ID NO: 1997)



GTCTTGGAGTGGCAGGTGGT



(SEQ ID NO: 1998)







MTMR11



TCTTCCGCTCAGTCTCCCAGT



(SEQ ID NO: 1999)



CTTCCGCTCAGTCTCCCAGT



(SEQ ID NO: 2000)



TTCCGCTCAGTCTCCCAGTC



(SEQ ID NO: 2001)



TGCCACCCACTCTCGCTGTA



(SEQ ID NO: 2002)



GCTCAGTCTCCCAGTCTTCC



(SEQ ID NO: 2003)







MYC



TCTTCCGCTCAGTCTCCCAGT



(SEQ ID NO: 2004)



CTTCCGCTCAGTCTCCCAGT  



(SEQ ID NO: 2005)



TGCCACCCACTCTCGCTGTA 



(SEQ ID NO: 2006)



TTCCGCTCAGTCTCCCAGTC 



(SEQ ID NO: 2007)



GCTCAGTCTCCCAGTCTTCC  



(SEQ ID NO: 2008)







NAT13



TCCACATTCTCCTCCTCCTCC



(SEQ ID NO: 2009)



CCACATTCTCCTCCTCCTCC 



(SEQ ID NO: 2010)



CCACATTCTCCTCCTCCTCCA



(SEQ ID NO: 2011)



CCTCTTCCACATTCTCCTCCT 



(SEQ ID NO: 2012)



CCTCTTCCACATTCTCCTCC  



(SEQ ID NO: 2013)







NCRNA00099



GTTCTCTCTTTCCCTGTCTC  



(SEQ ID NO: 2014)



TGTTCTCTCTTTCCCTGTCTC



(SEQ ID NO: 2015)



ACCCACTACATCACCTCCCA  



(SEQ ID NO: 2016)



CCCACTACATCACCTCCCA



(SEQ ID NO: 2017)



TGTTCTCTCTTTCCCTGTCT 



(SEQ ID NO: 2018)







NDUFB3



GTCCCTTCTATCTTCCATTGT



(SEQ ID NO: 2019)



TCCCTTCTATCTTCCATTGTC



(SEQ ID NO: 2020)



GTCCCTTCTATCTTCCATTG  



(SEQ ID NO: 2021)



GTGTCCCTTCTATCTTCCAT 



(SEQ ID NO: 2022)



GTCCCTTCTATCTTCCATT



(SEQ ID NO: 2023)







NDUFS5



CTCGTGCCCTTGACTCTTCTC 



(SEQ ID NO: 2024)



TCGTGCCCTTGACTCTTCTCT 



(SEQ ID NO: 2025)



GTGCCCTTGACTCTTCTCTG 



(SEQ ID NO: 2026)



CGTGCCCTTGACTCTTCTCT 



(SEQ ID NO: 2027)



CGTGCCCTTGACTCTTCTCTG 



(SEQ ID NO: 2028)







NFYA



CTCTCTCTCTCTTCCTCTCCC



(SEQ ID NO: 2029)



TCTCTCTCTCTTCCTCTCCC



(SEQ ID NO: 2030)



CTCTCTCTCTTCCTCTCCC



(SEQ ID NO: 2031)



CCCACCACCTTCCCACATTCT



(SEQ ID NO: 2032)



CCCACCACCTTCCCACATTC



(SEQ ID NO: 2033)







NPEPPS



GTAGCTCCACCTTATCCCTGC



(SEQ ID NO: 2034)



GCTCCACCTTATCCCTGCA



(SEQ ID NO: 2035)



AGCTCCACCTTATCCCTGCA



(SEQ ID NO: 2036)



TCCCACAACCAGAGCAACCC



A (SEQ ID NO: 2037)



TCCCACAACCAGAGCAACCC



(SEQ ID NO: 2038)







NQO1



ACTCCACCACCTCCCATCCT



(SEQ ID NO: 2039)



ACTCCACCACCTCCCATCCTT



(SEQ ID NO: 2040)



CTCCACCACCTCCCATCCTT



(SEQ ID NO: 2041)



CCACCACCTCCCATCCTTTCT



(SEQ ID NO: 2042)



CTCCACCACCTCCCATCCTTT



(SEQ ID NO: 2043)







NUAK2



CCTACTTCCCATATCCAGCCC



(SEQ ID NO: 2044)



GCGTTACTCCAGACCATTCCC



(SEQ ID NO: 2045)



CGCCTTCTTCACCTTCCCGT



(SEQ ID NO: 2046)



GCCTTCTTCACCTTCCCGT



(SEQ ID NO: 2047)



GCCTTCTTCACCTTCCCGTA



(SEQ ID NO: 2048)







PAX8



ACACCACACCTCACCTCCCA



(SEQ ID NO: 2049)



GCCCAGTCTTCTCTCTCCCT



(SEQ ID NO: 2050)



GCCCAGTCTTCTCTCTCCCTT



(SEQ ID NO: 2051)



CCCAGTCTTCTCTCTCCCTTC



(SEQ ID NO: 2052)



AGCCTCCCTCACCTTGTCCT



(SEQ ID NO: 2053)







PBLD



ACACCACACCTCACCTCCCA



(SEQ ID NO: 2054)



GCCCAGTCTTCTCTCTCCCT



(SEQ ID NO: 2055)



GCCCAGTCTTCTCTCTCCCTT



(SEQ ID NO: 2056)



CCCAGTCTTCTCTCTCCCTTC



(SEQ ID NO: 2057)



AGCCTCCCTCACCTTGTCCT



(SEQ ID NO: 2058)







PCBP2



GTCCCACCATTTATCTTCTCT



(SEQ ID NO: 2059)



GTCCCACCATTTATCTTCTC



(SEQ ID NO: 2060)



TCCCACCATTTATCTTCTCTG



(SEQ ID NO: 2061)



GTCCCACCATTTATCTTCT



(SEQ ID NO: 2062)



TGTCCCACCATTTATCTTCTC



(SEQ ID NO: 2063)







PDCD10



GTCTCTTCCTTCTTCCAGTGC



(SEQ ID NO: 2064)



TCCTTCTTCCAGTGCTCCTCT



(SEQ ID NO: 2065)



CCTTCTTCCAGTGCTCCTCT



(SEQ ID NO: 2066)



GTCTCTTCCTTCTTCCAGTG



(SEQ ID NO: 2067)



TCTTCCAGTGCTCCTCTTCC



(SEQ ID NO: 2068)







PKP1



CCATTGCCTGCCAGTTTCCC



(SEQ ID NO: 2069)



CCCAACTCCATCCCACTAGA



(SEQ ID NO: 2070)



ACTCCATCCCACTAGAAGCC



(SEQ ID NO: 2071)



ACTCCATCCCACTAGAAGCCG



(SEQ ID NO: 2072)



GGTTCTGGGTGGTCTTCTGG



(SEQ ID NO: 2073)







PLCB3



GCTCCTTCTTCTCCCTCTCGT



(SEQ ID NO: 2074)



GTCTGTTCCTCCTCTTCCTC



(SEQ ID NO: 2075)



CTCCTTCTTCTCCCTCTCGT



(SEQ ID NO: 2076)



GTCTGTCTGTTCCTCCTCTTC



(SEQ ID NO: 2077)



CTGTCTGTTCCTCCTCTTCCT



(SEQ ID NO: 2078)







POLH



GCCACCATTGTACCTGTTCCC 



(SEQ ID NO: 2079)



ATCCACTCACCTCAGCCTCC 



(SEQ ID NO: 2080)



GTCCCTGCTCCTGTTTCCA



(SEQ ID NO: 2081)



AGTCCCTGCTCCTGTTTCCA 



(SEQ ID NO: 2082)



TAGTCCACCCACCTCAGCCT 



(SEQ ID NO: 2083)







PPP1R10



GTTTCCAGTCTCTCTCCTCCC



(SEQ ID NO: 2084)



GATGCCCACTTCCCATGCCA 



(SEQ ID NO: 2085)



GCTTCTCCCTCTTCTTCTCT  



(SEQ ID NO: 2086)



ATGCCCACTTCCCATGCCAC  



(SEQ ID NO: 2087)



TTTCCAGTCTCTCTCCTCCC  



(SEQ ID NO: 2088)







PSIMCT-1



GTACCTCCTTTCCTCACTCGC 



(SEQ ID NO: 2089)



ACCTCCTTTCCTCACTCGC



(SEQ ID NO: 2090)



TACCTCCTTTCCTCACTCGC  



(SEQ ID NO: 2091)



GTACCTCCTTTCCTCACTCG 



(SEQ ID NO: 2092)



GTACCTCCTTTCCTCACTC



(SEQ ID NO: 2093)







PTRH2



GTACCTCCTTTCCTCACTCGC



(SEQ ID NO: 2094)



ACCTCCTTTCCTCACTCGC



(SEQ ID NO: 2095)



TACCTCCTTTCCTCACTCGC 



(SEQ ID NO: 2096)



GTACCTCCTTTCCTCACTCG 



(SEQ ID NO: 2097)



GTACCTCCTTTCCTCACTC



(SEQ ID NO: 2098)







PXN



CTTCACACCAGAGCCACCATC 



(SEQ ID NO: 2099)



TCACACCAGAGCCACCATCA 



G (SEQ ID NO: 2100)



ACTTCACACCAGAGCCACCA  



(SEQ ID NO: 2101)



TCACACCAGAGCCACCATCA 



(SEQ ID NO: 2102)



CACACCAGAGCCACCATCAG 



(SEQ ID NO: 2103)







QARS



GTCTCGCCATTCTCCACCAC



(SEQ ID NO: 2104)



GTCTCGCCATTCTCCACCACA



(SEQ ID NO: 2105)



GCCATTCTCCACCACATCCT 



(SEQ ID NO: 2106)



GTCTCGCCATTCTCCACCA



(SEQ ID NO: 2107)



GCCATTCTCCACCACATCCTT



(SEQ ID NO: 2108)







RABL3



GCCACTGCTTGCTCACTCTTC



(SEQ ID NO: 2109)



CCTCCCGTATTGTCACTTCCT



(SEQ ID NO: 2110)



AGTCCGCCTCTTCCAGTCAC



(SEQ ID NO: 2111)



CCTCCCGTATTGTCACTTCC



(SEQ ID NO: 2112)



GCCACTGCTTGCTCACTCTT



(SEQ ID NO: 2113)







RAD51L1



TTCCTGTTCCCTGTGTGCCC



(SEQ ID NO: 2114)



TTTCCTGTTCCCTGTGTGCCC



(SEQ ID NO: 2115)



TTTCCTGTTCCCTGTGTGCC



(SEQ ID NO: 2116)



TTCCTGTTCCCTGTGTGCC



(SEQ ID NO: 2117)



CCCATTTCCTGTTCCCTGTGT



(SEQ ID NO: 2118)







RBMX



TCCTCCACTTCCTCCTCTTCC



(SEQ ID NO: 2119)



CCTCCACTTCCTCCTCTTCC 



(SEQ ID NO: 2120)



CCTCCACTTCCTCCTCTTCCA



(SEQ ID NO: 2121)



CCACTTCCTCCTCTTCCACCT



(SEQ ID NO: 2122)



TCCACTTCCTCCTCTTCCACC



(SEQ ID NO: 2123)







RGD1308059



GCTCCCTACCGAGATGCCAT



(SEQ ID NO: 2124)



GTTCACCATCTCCTGCCTG



(SEQ ID NO: 2125)



GTTTGGTCTCTAGCCTCTCC



(SEQ ID NO: 2126)



GCAGTTTGGTCTCTAGCCTCT



(SEQ ID NO: 2127)



AGTTTGGTCTCTAGCCTCTCC



(SEQ ID NO: 2128)







RGD1309079



GCTCTTACCCTATGTCTTCCC



(SEQ ID NO: 2129)



ACAGCCACCATTCTTCCACA



(SEQ ID NO: 2130)



GCTCTCATACCATACATCCCA



(SEQ ID NO: 2131)



GCTGTCTTCCTCCACTTTG



(SEQ ID NO: 2132)



TGCTGTCTTCCTCCACTTTG



(SEQ ID NO: 2133)







RGD1309492



GCTCTTACCCTATGTCTTCCC



(SEQ ID NO: 2134)



ACAGCCACCATTCTTCCACA



(SEQ ID NO: 2135)



GCTCTCATACCATACATCCCA



(SEQ ID NO: 2136)



GCTGTCTTCCTCCACTTTG



(SEQ ID NO: 2137)



TGCTGTCTTCCTCCACTTTG



(SEQ ID NO: 2138)







MRI1



ACCTCTTCCCGCCCGATTT



(SEQ ID NO: 2139)



ACCTCTTCCCGCCCGATTT



(SEQ ID NO: 2140)



TTTACCTCTTCCCGCCCGA



(SEQ ID NO: 2141)



TACCTCTTCCCGCCCGATTT



(SEQ ID NO: 2142)



TTACCTCTTCCCGCCCGATT



(SEQ ID NO: 2143)







RNU86



AGGAGCAGAGCAGAGTTGGG



(SEQ ID NO: 2144)



TCAGGAGCAGAGCAGAGTTG



G (SEQ ID NO: 2145)



CTTCAGGAGCAGAGCAGAGT



T (SEQ ID NO: 2146)



CTTCAGGAGCAGAGCAGAGT



(SEQ ID NO: 2147)



CAGGAGCAGAGCAGAGTTGG



(SEQ ID NO: 2148)







RPL18AP3



GCGATCTCCTCCACCTTCAT



(SEQ ID NO: 2149)



GCGATCTCCTCCACCTTCA



(SEQ ID NO: 2150)



GCGATCTCCTCCACCTTCATG



(SEQ ID NO: 2151)



GGTGGTCAGGTCCCGGTATT



(SEQ ID NO: 2152)



GACCCACTACCTTGTACTCTC



(SEQ ID NO: 2153)







RPL27A



CTCTCCACCATAGCACTTCCC 



(SEQ ID NO: 2154)



CCTCCCATTGTAGCCGTCCT 



(SEQ ID NO: 2155)



CTCCACCATAGCACTTCCCGT



(SEQ ID NO: 2156)



CCTCCCATTGTAGCCGTCCTT



(SEQ ID NO: 2157)



TCTCCACCATAGCACTTCCC  



(SEQ ID NO: 2158)







RPL29P2



GCACCTGTCCTTCTGTCCTC  



(SEQ ID NO: 2159)



GCACCTGTCCTTCTGTCCTCA



(SEQ ID NO: 2160)



GGCACCTGTCCTTCTGTCCT  



(SEQ ID NO: 2161)



GCACCTGTCCTTCTGTCCT



(SEQ ID NO: 2162)



TGGCACCTGTCCTTCTGTCCT 



(SEQ ID NO: 2163)







RPL29P31



GGTTGTTTGTGCTGTGGTTCT



(SEQ ID NO: 2164)



GGTTGTTTGTGCTGTGGTTC  



(SEQ ID NO: 2165)



CTGGTTGTTTGTGCTGTGGT  



(SEQ ID NO: 2166)



CTGGTTGTTTGTGCTGTGGTT



(SEQ ID NO: 2167)



TGGTTGTTTGTGCTGTGGTTC 



(SEQ ID NO: 2168)







RPL3



GCCTCCACCACCTCCTTCTT  



(SEQ ID NO: 2169)



CCTCCACCACCTCCTTCTT



(SEQ ID NO: 2170)



AGCCTCCACCACCTCCTTCT  



(SEQ ID NO: 2171)



AGCCTCCACCACCTCCTTCTT



(SEQ ID NO: 2172)



GTTCCCACCACACAGCCTTTC 



(SEQ ID NO: 2173)







RPL34



GTGCTTTCCCAACCTTCTT



(SEQ ID NO: 2174)



GTGCTTTCCCAACCTTCTTG  



(SEQ ID NO: 2175)



GCTTTCTGACTCTGTGCTTGT 



(SEQ ID NO: 2176)



GGGTTCGGGACAGCCTAGTT 



(SEQ ID NO: 2177)



GGACCATTCTGAGTGCCTGC  



(SEQ ID NO: 2178)







RPL37A



GCTCGTCTCTTCATCTTGGT



(SEQ ID NO: 2179)



GCTCGTCTCTTCATCTTGGTT



(SEQ ID NO: 2180)



GAACCACAGTGCCAGATCCC



(SEQ ID NO: 2181)



AACCACAGTGCCAGATCCC



(SEQ ID NO: 2182)



CTCGTCTCTTCATCTTGGTT



(SEQ ID NO: 2183)







RPL4



GTCCCACCACCTCGAACTCT



(SEQ ID NO: 2184)



AGTCCCACCACCTCGAACTCT



(SEQ ID NO: 2185)



TCCCACCACCTCGAACTCTG



(SEQ ID NO: 2186)



TCCCACCACCTCGAACTCT



(SEQ ID NO: 2187)



AGTCCCACCACCTCGAACTC



(SEQ ID NO: 2188)







RPL7A



GTCCCAGTCTTGCCTTTCCCT



(SEQ ID NO: 2189)



GTCCCAGTCTTGCCTTTCCC



(SEQ ID NO: 2190)



GAGCCACCTTCTTTCCCTTG



(SEQ ID NO: 2191)



GAGCCACCTTCTTTCCCTT



(SEQ ID NO: 2192)



TCCCAGTCTTGCCTTTCCCT



(SEQ ID NO: 2193)







RPS18



AGTTCTCCCGCCCTCTTGGT



(SEQ ID NO: 2194)



CGTTCCACCTCATCCTCAG



(SEQ ID NO: 2195)



CGTTCCACCTCATCCTCAGT



(SEQ ID NO: 2196)



GTTCCACCTCATCCTCAGT



(SEQ ID NO: 2197)



TCAGTGAGTTCTCCCGCCCT



(SEQ ID NO: 2198)







RPSA



ACTTCCCGAGCCAGCATCCA 



(SEQ ID NO: 2199)



GAGCCAGCATCCACCACATC



A (SEQ ID NO: 2200)



GAGCCAGCATCCACCACATC



(SEQ ID NO: 2201)



AACTTCCCGAGCCAGCATCCA



(SEQ ID NO: 2202)



AACTTCCCGAGCCAGCATCC



(SEQ ID NO: 2203)







SDF2



CACCTCACCATCTCTCACCC



(SEQ ID NO: 2204)



CACCTCACCATCTCTCACCCA



(SEQ ID NO: 2205)



ACCTCACCATCTCTCACCCA



(SEQ ID NO: 2206)



TCACCTTCACCTTCCTCACCA



(SEQ ID NO: 2207)



TCACCTTCACCTTCCTCACC



(SEQ ID NO: 2208)







SEMA3C



GTCCTCTGATCTCCTCCCTCT



(SEQ ID NO: 2209)



GTCCTCTGATCTCCTCCCTC



(SEQ ID NO: 2210)



CCTCTGATCTCCTCCCTCTGT



(SEQ ID NO: 2211)



CGCCACTCCCACAGACATAC



A (SEQ ID NO: 2212)



GCCACTCCCACAGACATACA



(SEQ ID NO: 2213)







SERAC1



CCTTCCTTCTGTGCCTGCCA



(SEQ ID NO: 2214)



AGCCCTTCCTTCTGTGCCTG



(SEQ ID NO: 2215)



AGCCCTTCCTTCTGTGCCT



(SEQ ID NO: 2216)



CAGCCCTTCCTTCTGTGCCT



(SEQ ID NO: 2217)



ATCTCTCCAGTGTGTGCCA



(SEQ ID NO: 2218)







SERPINI1



GCCTCTCCCTCCTCTAGTTG



(SEQ ID NO: 2219)



GCCTCTCCCTCCTCTAGTT



(SEQ ID NO: 2220)



CGCCTCTCCCTCCTCTAGTT



(SEQ ID NO: 2221)



GTTCCTGCTTTCGTCTCTCCC



(SEQ ID NO: 2222)



TTCCTGCTTTCGTCTCTCCC



(SEQ ID NO: 2223)







SF3B4



GGTGGGAATGGGTGAGGATG



T (SEQ ID NO: 2224)



GTTCACCCGTATTGGCTTCCC



(SEQ ID NO: 2225)



GCCAGTGACTCTATCCTTTGG



(SEQ ID NO: 2226)



CAGGATTGGGAGCAGAGGGT



(SEQ ID NO: 2227)



TGGGTGAGGATGTGAGTGT



(SEQ ID NO: 2228)







SFRS3



AGTCTTCCCGCTTTCCTCCG  



(SEQ ID NO: 2229)



AGTCTTCCCGCTTTCCTCC



(SEQ ID NO: 2230)



AGCCACCACATCCAGCCAGT 



(SEQ ID NO: 2231)



ACCAACTCTCTCACTCACCC  



(SEQ ID NO: 2232)



GAGTCTTCCCGCTTTCCTCC  



(SEQ ID NO: 2233)







SFXN1



CCTCCCTGCTTTACAATCCCT 



(SEQ ID NO: 2234)



TCCTCCCTGCTTTACAATCCC



(SEQ ID NO: 2235)



CCTCCCTGCTTTACAATCCC 



(SEQ ID NO: 2236)



GTTCCCATTCTCATCCGTG



(SEQ ID NO: 2237)



GTTTCCTCCCTGCTTTACA



(SEQ ID NO: 2238)







SKIL



CTGCCATCATCTCCCATCCC 



(SEQ ID NO: 2239)



GTCTTGCTTCCCGTTCCTGTC 



(SEQ ID NO: 2240)



GCCATCATCTCCCATCCCAT 



(SEQ ID NO: 2241)



GCCATCATCTCCCATCCCA



(SEQ ID NO: 2242)



GCCATCATCTCCCATCCCATT



(SEQ ID NO: 2243)







SLC25A25



TTCCTCCCTCTCTCTCCAC



(SEQ ID NO: 2244)



GTTCCTCCCTCTCTCTCCAC  



(SEQ ID NO: 2245)



CCTCCTCCCACCTTACCTCA  



(SEQ ID NO: 2246)



GTTCCTCCCTCTCTCTCCA



(SEQ ID NO: 2247)



CCTTCTCCTCCTCCTCCCAT 



(SEQ ID NO: 2248)







SLC38A2



GTTGCCACAGTATCTTCTCCC



(SEQ ID NO: 2249)



CTGTCTTCCCACTGCCTTTCT



(SEQ ID NO: 2250)



GCCACAGTATCTTCTCCCAT 



(SEQ ID NO: 2251)



TCTGTCTTCCCACTGCCTTTC



(SEQ ID NO: 2252)



GCCACAGTATCTTCTCCCA



(SEQ ID NO: 2253)







SLC39A14



TCCTGTCTCCTGTCTCTTCT



(SEQ ID NO: 2254)



CTTCCTGTCTCCTGTCTCTTC



(SEQ ID NO: 2255)



TTCCTGTCTCCTGTCTCTTCT



(SEQ ID NO: 2256)



GCTTCCTGTCTCCTGTCTCT



(SEQ ID NO: 2257)



CTTCCTGTCTCCTGTCTCTT



(SEQ ID NO: 2258)







SMC6



GTCCCTCTAGCCTGTGTCCT



(SEQ ID NO: 2259)



AGTCCCTCTAGCCTGTGTCCT



(SEQ ID NO: 2260)



TCCCTCTAGCCTGTGTCCTCT



(SEQ ID NO: 2261)



AGTCCCTCTAGCCTGTGTCC 



(SEQ ID NO: 2262)



TCCCTCTAGCCTGTGTCCTC 



(SEQ ID NO: 2263)







SNORA1



GATCAACAGTGCCCATTCTCT



(SEQ ID NO: 2264)



ATCAACAGTGCCCATTCTCT



(SEQ ID NO: 2265)



TCAACAGTGCCCATTCTCTAG



(SEQ ID NO: 2266)



TCAACAGTGCCCATTCTCT



(SEQ ID NO: 2267)



TCAACAGTGCCCATTCTCTA 



(SEQ ID NO: 2268)







SNORA18



GTCTTGTAATTCCTTCCCAC 



(SEQ ID NO: 2269)



CTGTCTTGTAATTCCTTCCC



(SEQ ID NO: 2270)



GTAATTCCTTCCCACAGATC



(SEQ ID NO: 2271)



GTCTTGTAATTCCTTCCCACA



(SEQ ID NO: 2272)



GTCTTGTAATTCCTTCCCA



(SEQ ID NO: 2273)







SNORA19



CTGAAGGAGACTGGCAGCAT 



T (SEQ ID NO: 2274)



CTGAAGGAGACTGGCAGCAT 



(SEQ ID NO: 2275)



CTGAAGGAGACTGGCAGCA



(SEQ ID NO: 2276)



GAAGGAGACTGGCAGCATTA 



C (SEQ ID NO: 2277)



AGGAGACTGGCAGCATTAC



(SEQ ID NO: 2278)







SNORA3



GGACTCTAGCACACTCAGGC



A (SEQ ID NO: 2279)



GGACTCTAGCACACTCAGGC



(SEQ ID NO: 2280)



AGGACTCTAGCACACTCAGG



C (SEQ ID NO: 2281)



GGACTCTAGCACACTCAGG



(SEQ ID NO: 2282)



GACTCTAGCACACTCAGGCA



(SEQ ID NO: 2283)







SNORA32



GTCATGTCCACAGCAAATG



(SEQ ID NO: 2284)



GTCATGTCCACAGCAAATGA



G (SEQ ID NO: 2285)



GTCATGTCCACAGCAAATGA



(SEQ ID NO: 2286)



CATGTCCACAGCAAATGAGA



(SEQ ID NO: 2287)



TCATGTCCACAGCAAATGAG



A (SEQ ID NO: 2288)







SNORA38



GGGAACCACAGACACGCCTT



(SEQ ID NO: 2289)



GGGAACCACAGACACGCCTT



T (SEQ ID NO: 2290)



GCTGGCCTCAAAGTTTCCCA



(SEQ ID NO: 2291)



AGCTGGCCTCAAAGTTTCCC



(SEQ ID NO: 2292)



AGCTGGCCTCAAAGTTTCCCA



(SEQ ID NO: 2293)







SNORA40



TCTGTAAGTCTTTGTCCACGT



(SEQ ID NO: 2294)



CCTATCTGTAAGTCTTTGTCC



(SEQ ID NO: 2295)



CTGTAAGTCTTTGTCCACGTT



(SEQ ID NO: 2296)



CTGTAAGTCTTTGTCCACGT



(SEQ ID NO: 2297)



TCTGTAAGTCTTTGTCCACG



(SEQ ID NO: 2298)







SNORA45



GTACTCTAGCACACCAGGAC



A (SEQ ID NO: 2299)



GTACTCTAGCACACCAGGAC



(SEQ ID NO: 2300)



GTATGTTTCTGATCTGGGCAC



(SEQ ID NO: 2301)



GTATGTTTCTGATCTGGGCA



(SEQ ID NO: 2302)



ATGTTTCTGATCTGGGCACT



(SEQ ID NO: 2303)







SNORA54



AGCCACCAGTCAGTCAGTCA  



(SEQ ID NO: 2304)



GCCACCAGTCAGTCAGTCA



(SEQ ID NO: 2305)



GCCACCAGTCAGTCAGTCAT  



(SEQ ID NO: 2306)



GCCACCAGTCAGTCAGTCATG



(SEQ ID NO: 2307)



AGCCACCAGTCAGTCAGTCAT 



(SEQ ID NO: 2308)







SNORA6



GCACACTTCCGACACTCAG



(SEQ ID NO: 2309)



AGCACACTTCCGACACTCAG  



(SEQ ID NO: 2310)



GCCTCCACCCTGAGCTTTA



(SEQ ID NO: 2311)



TAGCACACTTCCGACACTCAG 



(SEQ ID NO: 2312)



AGCACACTTCCGACACTCA



(SEQ ID NO: 2313)







SNORA62



GCCAGTCAGTAGCCTCAACTC



(SEQ ID NO: 2314)



GCCAGTCAGTAGCCTCAACT  



(SEQ ID NO: 2315)



GCCAGTCAGTAGCCTCAAC



(SEQ ID NO: 2316)



CCAGTCAGTAGCCTCAACTCC 



(SEQ ID NO: 2317)



AGTCAGTAGCCTCAACTCCA  



(SEQ ID NO: 2318)







SNORA76



TCTAGCGCTGCAGATTGACCC



(SEQ ID NO: 2319)



CTAGCGCTGCAGATTGACCC  



(SEQ ID NO: 2320)



GCACATGCTCTAGCGCTGCA  



(SEQ ID NO: 2321)



TAGCGCTGCAGATTGACCC



(SEQ ID NO: 2322)



TCTAGCGCTGCAGATTGACC 



(SEQ ID NO: 2323)







SNORA8



CTGACCTTTGAACACCCTAGC



(SEQ ID NO: 2324)



GCACTGACCTTTGAACACCCT



(SEQ ID NO: 2325)



GACCTTTGAACACCCTAGCAG



(SEQ ID NO: 2326)



GACCTTTGAACACCCTAGCA 



(SEQ ID NO: 2327)



TGACCTTTGAACACCCTAGCA 



(SEQ ID NO: 2328)







SNORA84



AGCATCCAGCAACCACAGGG



(SEQ ID NO: 2329)



ACAGCATCCAGCAACCACAG 



G (SEQ ID NO: 2330)



CAGCATCCAGCAACCACAGG



(SEQ ID NO: 2331)



AGCATCCAGCAACCACAGG



(SEQ ID NO: 2332)



ACAGCATCCAGCAACCACAG



(SEQ ID NO: 2333)







SNORD16



TTTCGTCAACCTTCTGTACCA



(SEQ ID NO: 2334)



TTCGTCAACCTTCTGTACCA



(SEQ ID NO: 2335)



TCGTCAACCTTCTGTACCA



(SEQ ID NO: 2336)



TTTCGTCAACCTTCTGTACC



(SEQ ID NO: 2337)



TTCGTCAACCTTCTGTACC



(SEQ ID NO: 2338)







SNORD18A



ACACGGACCAATGAAGTGG



(SEQ ID NO: 2339)



AACACGGACCAATGAAGTGG



(SEQ ID NO: 2340)



AAACACGGACCAATGAAGTG 



G (SEQ ID NO: 2341)



GTTCAGAAACACGGACCAAT



G (SEQ ID NO: 2342)



AGAAACACGGACCAATGAAG



T (SEQ ID NO: 2343)







SNORD18B



GTTTCAGAAACACGGACCA



(SEQ ID NO: 2344)



GTTTCAGAAACACGGACCAA



T (SEQ ID NO: 2345)



GTTTCAGAAACACGGACCAA



(SEQ ID NO: 2346)



TTTCAGAAACACGGACCAA



(SEQ ID NO: 2347)



TCAGAAACACGGACCAATT



(SEQ ID NO: 2348)







SNORD18C



GACCTTAAGTGGAATCTCATC



(SEQ ID NO: 2349)



CGGACCTTAAGTGGAATCTCA



(SEQ ID NO: 2350)



ACGGACCTTAAGTGGAATCTC



(SEQ ID NO: 2351)



GGACCTTAAGTGGAATCTCAT



(SEQ ID NO: 2352)



GGACCTTAAGTGGAATCTCA



(SEQ ID NO: 2353)







SNORD24



TGTCATCACCATCTCTCAG



(SEQ ID NO: 2354)



TCAGCGATCTTGGTGGTTT



(SEQ ID NO: 2355)



TCAGCGATCTTGGTGGTTTA



(SEQ ID NO: 2356)



GCATCAGCGATCTTGGTGGTT



(SEQ ID NO: 2357)



GCATCAGCGATCTTGGTGGT



(SEQ ID NO: 2358)







SNORD35B



CTTTCTCAGCAGTGCCCACGT



(SEQ ID NO: 2359)



TTTCTCAGCAGTGCCCACGT



(SEQ ID NO: 2360)



TTCTCAGCAGTGCCCACGT



(SEQ ID NO: 2361)



CTTTCTCAGCAGTGCCCACG



(SEQ ID NO: 2362)



CTTTCTCAGCAGTGCCCAC



(SEQ ID NO: 2363)







SNORD36A



CATGGATTCGCACTTCAAGGT



(SEQ ID NO: 2364)



GGATTCGCACTTCAAGGTTG



(SEQ ID NO: 2365)



TGGATTCGCACTTCAAGGTTG



(SEQ ID NO: 2366)



CTCATGGATTCGCACTTCA



(SEQ ID NO: 2367)



ATGGATTCGCACTTCAAGGTT



(SEQ ID NO: 2368)







SNORD36B



CTTCACAGTAATTTCAGGC



(SEQ ID NO: 2369)



CTTCACAGTAATTTCAGGCCA



(SEQ ID NO: 2370)



CTTCACAGTAATTTCAGGCC



(SEQ ID NO: 2371)



TCACAGTAATTTCAGGCCA



(SEQ ID NO: 2372)



TCACAGTAATTTCAGGCCAAG



(SEQ ID NO: 2373)







SNORD36C



GCCTCAGATGCAATGCTGACC



(SEQ ID NO: 2374)



CAGATGCAATGCTGACCACA



(SEQ ID NO: 2375)



CAGATGCAATGCTGACCAC



(SEQ ID NO: 2376)



CAGATGCAATGCTGACCACA



T (SEQ ID NO: 2377)



CCACATGGTTTATTCAGGT



(SEQ ID NO: 2378)







SNORD3B-2



TTCTCTCCCTCTCACTCCC



(SEQ ID NO: 2379)



GTTCTCTCCCTCTCACTCCC 



(SEQ ID NO: 2380)



CGTTCTCTCCCTCTCACTCC 



(SEQ ID NO: 2381)



GTTCTCTCCCTCTCACTCC



(SEQ ID NO: 2382)



GCGTTCTCTCCCTCTCACTC 



(SEQ ID NO: 2383)







SNORD43



AGTTTCTGTCCGCCCGTCA



(SEQ ID NO: 2384)



GTTTCTGTCCGCCCGTCAAT  



(SEQ ID NO: 2385)



GTTTCTGTCCGCCCGTCAA



(SEQ ID NO: 2386)



AGTTTCTGTCCGCCCGTCAA  



(SEQ ID NO: 2387)



AGTTTCTGTCCGCCCGTCAAT 



(SEQ ID NO: 2388)







SNORD44



ACCTTCATGTTCAGTCAGCA 



(SEQ ID NO: 2389)



ACCTTCATGTTCAGTCAGCAT



(SEQ ID NO: 2390)



CCTTCATGTTCAGTCAGCAT 



(SEQ ID NO: 2391)



CCTTCATGTTCAGTCAGCATT 



(SEQ ID NO: 2392)



CCTTCATGTTCAGTCAGCA



(SEQ ID NO: 2393)







SNORD5



CCCGTTATCATTCAGCAGTTG



(SEQ ID NO: 2394)



CCCGTTATCATTCAGCAGTT 



(SEQ ID NO: 2395)



CCCGTTATCATTCAGCAGT



(SEQ ID NO: 2396)



GCCCGTTATCATTCAGCAGT  



(SEQ ID NO: 2397)



GCCCGTTATCATTCAGCAGTT



(SEQ ID NO: 2398)







SNORD58A



CCCGTTATCATTCAGCAGTTG



(SEQ ID NO: 2399)



CCCGTTATCATTCAGCAGTT 



(SEQ ID NO: 2400)



CCCGTTATCATTCAGCAGT



(SEQ ID NO: 2401)



GCCCGTTATCATTCAGCAGT  



(SEQ ID NO: 2402)



GCCCGTTATCATTCAGCAGTT



(SEQ ID NO: 2403)







SNORD6



ACATTTCGCCCATCATCAT



(SEQ ID NO: 2404)



CATTTCGCCCATCATCATA



(SEQ ID NO: 2405)



AACATTTCGCCCATCATCA



(SEQ ID NO: 2406)



TTCGCCCATCATCATAACATC



(SEQ ID NO: 2407)



GAGCAGTTGAACATTTCGCCC



(SEQ ID NO: 2408)







SNORD60



GAGGTGTCAGAAGTCAAAGC 



A (SEQ ID NO: 2409)



CATACGAGGTGTCAGAAGTC



A (SEQ ID NO: 2410)



CGAGGTGTCAGAAGTCAAAG 



C (SEQ ID NO: 2411)



GAGGTGTCAGAAGTCAAAGC



(SEQ ID NO: 2412)



CATACGAGGTGTCAGAAGTC



(SEQ ID NO: 2413)







SNORD61



TCATGTCAGACGATCAATG



(SEQ ID NO: 2414)



TCATGTCAGACGATCAATGCA



(SEQ ID NO: 2415)



ATCATGTCAGACGATCAATGC



(SEQ ID NO: 2416)



CATGTCAGACGATCAATGCA



(SEQ ID NO: 2417)



GTCAGACGATCAATGCAATC



A (SEQ ID NO: 2418)







SNORD74



GTCAGATGTCCCTACCAAC



(SEQ ID NO: 2419)



ACTGTCAGATGTCCCTACC



(SEQ ID NO: 2420)



TTACTGTCAGATGTCCCTACC



(SEQ ID NO: 2421)



TACTGTCAGATGTCCCTACC



(SEQ ID NO: 2422)



ACACAGGCTTCATCAGAGG



(SEQ ID NO: 2423)







SNORD75



TCCCTTCTGTCCACTACTCT 



(SEQ ID NO: 2424)



TCCCTTCTGTCCACTACTCTT



(SEQ ID NO: 2425)



ATCCCTTCTGTCCACTACTCT



(SEQ ID NO: 2426)



ATCCCTTCTGTCCACTACTC



(SEQ ID NO: 2427)



TCCCTTCTGTCCACTACTC



(SEQ ID NO: 2428)







SNORD76



TCCTCATCATTCTAGCACTCA



(SEQ ID NO: 2429)



TCCTCATCATTCTAGCACTC



(SEQ ID NO: 2430)



ATCCTCATCATTCTAGCACTC



(SEQ ID NO: 2431)



CCTCATCATTCTAGCACTCA



(SEQ ID NO: 2432)



CCTCATCATTCTAGCACTC



(SEQ ID NO: 2433)







SNORD77



GCAACCATCATAGTATCTG



(SEQ ID NO: 2434)



TCTGCTGAACTATGCAACCAT



(SEQ ID NO: 2435)



ATCTGCTGAACTATGCAACCA



(SEQ ID NO: 2436)



TCTGCTGAACTATGCAACCA



(SEQ ID NO: 2437)



ATCTGCTGAACTATGCAACC



(SEQ ID NO: 2438)







SNORD78



GGTCAGACATTTGATCAAC



(SEQ ID NO: 2439)



CAGGTCAGACATTTGATCAAC



(SEQ ID NO: 2440)



CAGGTCAGACATTTGATCA



(SEQ ID NO: 2441)



TCAGGTCAGACATTTGATC



(SEQ ID NO: 2442)



GCTCATTTCAGGTCAGACATT



(SEQ ID NO: 2443)







SNORD80



CTCATCAGCGTTAGTCTGC



(SEQ ID NO: 2444)



GCTCATCAGCGTTAGTCTGC



(SEQ ID NO: 2445)



GCTCATCAGCGTTAGTCTGCT



(SEQ ID NO: 2446)



CATCAGATAGGAGCGAAAGA



C (SEQ ID NO: 2447)



GCGTTAGTCTGCTGAACTATG



(SEQ ID NO: 2448)







SNORD81



GAGAGAGTTCAAGTTGGATT



G (SEQ ID NO: 2449)



CAGTGAGAGAGTTCAAGTTG



G (SEQ ID NO: 2450)



AGTGAGAGAGTTCAAGTTGG



A (SEQ ID NO: 2451)



GTGAGAGAGTTCAAGTTGGA



T (SEQ ID NO: 2452)



GAGAGAGTTCAAGTTGGAT



(SEQ ID NO: 2453)







SNORD83A



GGAAGGCAGTAGAGAATGGT  



T (SEQ ID NO: 2454)



AGGAAGGCAGTAGAGAATGG 



T (SEQ ID NO: 2455)



GGAAGGCAGTAGAGAATGGT  



(SEQ ID NO: 2456)



CTCAGAAGGAAGGCAGTAGA  



(SEQ ID NO: 2457)



TCAGAAGGAAGGCAGTAGAG 



A (SEQ ID NO: 2458)







SNORD83B



ACGGTTCCGCAGTCTGTCTC  



(SEQ ID NO: 2459)



CTCAGAAGGAAGGCAACAAG 



G (SEQ ID NO: 2460)



AGGAAGGCAACAAGGAACGG 



(SEQ ID NO: 2461)



AGGAAGGCAACAAGGAACGG  



T (SEQ ID NO: 2462)



TCAGAAGGAAGGCAACAAGG 



A (SEQ ID NO: 2463)







SPATA19



CATCTTCTCCCTTACACCCT  



(SEQ ID NO: 2464)



ACATCTTCTCCCTTACACCCT



(SEQ ID NO: 2465)



ACATCTTCTCCCTTACACCC  



(SEQ ID NO: 2466)



CTTCTTGGCTCCCTCCTTCC 



(SEQ ID NO: 2467)



ATCTTCTCCCTTACACCCT



(SEQ ID NO: 2468)







SRP54



GTCGTCAACTCCCACCAGCT 



(SEQ ID NO: 2469)



CCCACAACCCAACTTCCCGA 



(SEQ ID NO: 2470)



GTCCTGCCATACCACCCATG  



(SEQ ID NO: 2471)



AGTCCTGCCATACCACCCAT  



(SEQ ID NO: 2472)



AGTCCTGCCATACCACCCA



(SEQ ID NO: 2473)







ST6GAL2



GTCGTCAACTCCCACCAGCT 



(SEQ ID NO: 2474)



CCCACAACCCAACTTCCCGA  



(SEQ ID NO: 2475)



GTCCTGCCATACCACCCATG  



(SEQ ID NO: 2476)



AGTCCTGCCATACCACCCAT  



(SEQ ID NO: 2477)



AGTCCTGCCATACCACCCA



(SEQ ID NO: 2478)







SUPT6H



CCTTCCTCTTCCTCCTCCCT



(SEQ ID NO: 2479)



ACCTTCCTCTTCCTCCTCCCT



(SEQ ID NO: 2480)



ACCTTCCTCTTCCTCCTCCC



(SEQ ID NO: 2481)



CTTCCTCTTCCTCCTCCCTG



(SEQ ID NO: 2482)



CTTCCTCTTCCTCCTCCCT



(SEQ ID NO: 2483)







TAF1



GCTCCTGTTCCTCCCGTTCA



(SEQ ID NO: 2484)



GTCTCCTTCTTCATCACTCCC



(SEQ ID NO: 2485)



TCTCCTTCTTCATCACTCCCA



(SEQ ID NO: 2486)



TCTCCTTCTTCATCACTCCC



(SEQ ID NO: 2487)



CTCCTCCTCCTGTTCTTCTGT



(SEQ ID NO: 2488)







TAF1D



TCAGCCCTCCAACTCCCGAT



(SEQ ID NO: 2489)



TCAGCCCTCCAACTCCCGATA



(SEQ ID NO: 2490)



CAGCCCTCCAACTCCCGATA



(SEQ ID NO: 2491)



CCTCTGCTGTTGACTCCTC



(SEQ ID NO: 2492)



AGCCCTCCAACTCCCGATA



(SEQ ID NO: 2493)







TBC1D5



CCTCTTTCTCTCTCTCTCCCT



(SEQ ID NO: 2494)



TCCTCTTTCTCTCTCTCTCCC



(SEQ ID NO: 2495)



CCTCTTTCTCTCTCTCTCCC



(SEQ ID NO: 2496)



GTGTCCCTCTGCCATCCTCA



(SEQ ID NO: 2497)



CTCTTTCTCTCTCTCTCCCTC



(SEQ ID NO: 2498)







TEX2



ACTCCTCCTCCTCTTCCTCCT



(SEQ ID NO: 2499)



ACTCCTCCTCCTCTTCCTCC



(SEQ ID NO: 2500)



CTCCTCTTCCTCCTCCTCCT



(SEQ ID NO: 2501)



CTCCTCCTCTTCCTCCTCCT



(SEQ ID NO: 2502)



CTCCTCCTCCTCTTCCTCCT 



(SEQ ID NO: 2503)







TEX21



CCCTCTCCTCTGGTCTTGCT



(SEQ ID NO: 2504)



ACCCTTCACCAGCCTCTTCA



(SEQ ID NO: 2505)



TGGTGTCTGGCTTCTGGTGG



(SEQ ID NO: 2506)



CCCTTCACCAGCCTCTTCA



(SEQ ID NO: 2507)



CCCTTCACCAGCCTCTTCAT



(SEQ ID NO: 2508)







TMEM49



TCCCGCTCCCTCCTCTTCTT



(SEQ ID NO: 2509)



TTCCCGCTCCCTCCTCTTCT



(SEQ ID NO: 2510)



TCTTCCCGCTCCCTCCTCTT



(SEQ ID NO: 2511)



TTCTTCCCGCTCCCTCCTCT



(SEQ ID NO: 2512)



TCCCGCTCCCTCCTCTTCTTT



(SEQ ID NO: 2513)







TNPO1



TTATTCCTCCCTCCCTGCC



(SEQ ID NO: 2514)



TGTTCCTCTTTCTTGTCCCT



(SEQ ID NO: 2515)



GTTCCTCTTTCTTGTCCCT



(SEQ ID NO: 2516)



TATTCCTCCCTCCCTGCCCA



(SEQ ID NO: 2516)



TTTATTCCTCCCTCCCTGCCC



(SEQ ID NO: 2517)







TRAF7



TTTCCTCTCTCCTCCTGCCC



(SEQ ID NO: 2518)



GTCATTCTCGCCCTCCCTTT



(SEQ ID NO: 2519)



CTCCTCCGTGCTTCTCTCCT



(SEQ ID NO: 2520)



GTCATTCTCGCCCTCCCTT



(SEQ ID NO: 2521)



TTTCCTCTCTCCTCCTGCC



(SEQ ID NO: 2522)







TRIM66



GTTCTCAGCCATTCTCCCACC



(SEQ ID NO: 2523)



CTTCCTCCCTACCCAACTCCT



(SEQ ID NO: 2524)



TCTCAGCCATTCTCCCACCAC



(SEQ ID NO: 2525)



TCTCAGCCATTCTCCCACCA



(SEQ ID NO: 2526)



TCTTCCTCCCTACCCAACTCC



(SEQ ID NO: 2527)







TSGA13



AGGGAGGTGGGTTGTTGGT



(SEQ ID NO: 2528)



CAGGGAGGTGGGTTGTTGGT  



(SEQ ID NO: 2529)



ACTCTCCTTGTCTTGCTGGGT



(SEQ ID NO: 2530)



GGTTCTGGTGGGCATGTCTTC



(SEQ ID NO: 2531)



AGGGAGGTGGGTTGTTGGTC  



(SEQ ID NO: 2532)







TUBD1



CACATTTCTCCACTTCCTTCC 



(SEQ ID NO: 2533)



ACTCCATTCTCCTCCTCACT  



(SEQ ID NO: 2534)



ACATTTCTCCACTTCCTTCC 



(SEQ ID NO: 2535)



CATTTCTCCACTTCCTTCC



(SEQ ID NO: 2536)



ACTCCATTCTCCTCCTCAC



(SEQ ID NO: 2537)







TYW3



CTCTTTCCCACCGTTATGCC  



(SEQ ID NO: 2538)



TTTCCTCTCTTTCCCACCG



(SEQ ID NO: 2539)



TTCCTCTCTTTCCCACCGTT 



(SEQ ID NO: 2540)



TTTCCTCTCTTTCCCACCGT  



(SEQ ID NO: 2541)



TCCCACCGTTATGCCAGAGT  



(SEQ ID NO: 2542)







U58



GGTGTCCTAAGATAGTCATC  



(SEQ ID NO: 2543)



GTGTCCTAAGATAGTCATC



(SEQ ID NO: 2544)



GGTGTCCTAAGATAGTCAT



(SEQ ID NO: 2545)



GGTGTCCTAAGATAGTCATCA 



(SEQ ID NO: 2546)



GTGTCCTAAGATAGTCATCAC 



(SEQ ID NO: 2547)







UBA52



CACTCTTCAGCCACCTCCCT  



(SEQ ID NO: 2548)



ACTCTTCAGCCACCTCCCT



(SEQ ID NO: 2549)



ACTCTTCAGCCACCTCCCTG 



(SEQ ID NO: 2550)



ATCCCTGTTACTCCCTCCC



(SEQ ID NO: 2551)



TATCCCTGTTACTCCCTCCC 



(SEQ ID NO: 2552)







USP10



TCCCATTCATCCTCGCTTCCT



(SEQ ID NO: 2553)



TCCCATTCATCCTCGCTTCC



(SEQ ID NO: 2554)



TTCCCATTCATCCTCGCTTCC



(SEQ ID NO: 2555)



GTTCCCATTCATCCTCGCTTC



(SEQ ID NO: 2556)



CTCTCGGGTTTGGTTGGGTC



(SEQ ID NO: 2557)







VOF16



CCTCCCAGTCCTTCTCTCCT



(SEQ ID NO: 2558)



AGCCTCCCAGTCCTTCTCTC



(SEQ ID NO: 2559)



CCTCCCAGTCCTTCTCTCCTA



(SEQ ID NO: 2560)



AGCCTCCCAGTCCTTCTCT



(SEQ ID NO: 2561)



TCCTTCCTTCCTTCCTTCCT 



(SEQ ID NO: 2562)







WDR51B



CCTCTTCACCCTTGTCTGGC



(SEQ ID NO: 2563)



GCCTCTTCACCCTTGTCTGG 



(SEQ ID NO: 2564)



CCTCTTCACCCTTGTCTGGCA



(SEQ ID NO: 2565)



GGGATGTGGTGTTCTTGGGT



(SEQ ID NO: 2566)



CTGGTTTCTGTTGTTGTGGT



(SEQ ID NO: 2567)







WDR81



GTCCTCCTCCTCCTCTTCCT



(SEQ ID NO: 2568)



TCCTTCTTCCTTCCCGTCCC 



(SEQ ID NO: 2569)



CTCCTCCTCCTCTTCCTCCT 



(SEQ ID NO: 2570)



TCCTCCTCCTCTTCCTCCTC



(SEQ ID NO: 2571)



TCCTCCTCCTCCTCTTCCTC



(SEQ ID NO: 2572)







WDR82



GTCCTTCTGTATCTGTCCCT



(SEQ ID NO: 2573)



CCCTCAGCATCTCTTCCACTC



(SEQ ID NO: 2574)



GTCCTTCTGTATCTGTCCCTT



(SEQ ID NO: 2575)



GTTCGTCCTCACAGCCACCT 



(SEQ ID NO: 2576)



CCTCCTCCTCTTCTTCCTGCT



(SEQ ID NO: 2577)







WIPF2



CCTATCTTCCTATCTCCCACC



(SEQ ID NO: 2578)



CCCTATCTTCCTATCTCCCAC



(SEQ ID NO: 2579)



ACCATTCTCCACCCTGCCCT



(SEQ ID NO: 2580)



CCATTCTCCACCCTGCCCTA



(SEQ ID NO: 2581)



ACCATTCTCCACCCTGCCCTA



(SEQ ID NO: 2582)







ZBTB37



CCCAAGCCACTCCTCCAGAT



(SEQ ID NO: 2583)



TCCACATAGCCACTTACTCCC



(SEQ ID NO: 2584)



CCCAAGCCACTCCTCCAGATT



(SEQ ID NO: 2585)



CCACATAGCCACTTACTCCCA



(SEQ ID NO: 2586)



GCCACTTACTCCCATCATTG



(SEQ ID NO: 2587)







ZC3H4



CCTCCACCTCCTTCTCATCCT



(SEQ ID NO: 2588)



TCCTCCACCTCCTTCTCATCC



(SEQ ID NO: 2589)



CCTCCACCTCCTTCTCATCC



(SEQ ID NO: 2590)



CTCCACCTCCTTCTCATCCTC



(SEQ ID NO: 2591)



TCTCTCCTCCTCCTCCTGCT



(SEQ ID NO: 2592)







ZHX2



TCTGCCGTCTGTCACCTCTCT



(SEQ ID NO: 2593)



TCTCCACCCTTAGCCGATCC



(SEQ ID NO: 2594)



TCTCCACCCTTAGCCGATCCA



(SEQ ID NO: 2595)



GTCTCCACCCTTAGCCGATC



(SEQ ID NO: 2596)



CCGTCTGTCACCTCTCTCCA



(SEQ ID NO: 2597)







ZMYND8



GCTTCTCCCTGTGTCCGTCT



(SEQ ID NO: 2598)



ACTCCCTGCTTGTCCCGTCT



(SEQ ID NO: 2599)



CTCCCTGCTTGTCCCGTCTT



(SEQ ID NO: 2600)



ACTCCCTGCTTGTCCCGTCTT



(SEQ ID NO: 2601)



CTTCTCCCTGTGTCCGTCTG



(SEQ ID NO: 2602)







ZNF143



GTCTTTCTCCTGTGTGTGTCC 



(SEQ ID NO: 2603)



TCTTTCTCCTGTGTGTGTCCT



(SEQ ID NO: 2604)



GCCTTCTCTCCACTCTGTTG 



(SEQ ID NO: 2605)



GCCTTCTCTCCACTCTGTT



(SEQ ID NO: 2606)



TGCCTTCTCTCCACTCTGTT  



(SEQ ID NO: 2607)







ZNF662



GCCCTTCCCACAGTCCTTAC



(SEQ ID NO: 2608)



GCCCTTCCCACAGTCCTTACA



(SEQ ID NO: 2609)



GCCCTTCCCACATTCCTTACA



(SEQ ID NO: 2610)



GCCCTTCCCACATTCCTTAC 



(SEQ ID NO: 2611)



CCCTTCCCACATTCCTTACA



(SEQ ID NO: 2612)







ZWILCH



GTCTGCTCCCTCTGTGTCCT



(SEQ ID NO: 2613)



TGTCTGCTCCCTCTGTGTCCT



(SEQ ID NO: 2614)



TCTGCTCCCTCTGTGTCCTC



(SEQ ID NO: 2615)



TGTCTGCTCCCTCTGTGTCC



(SEQ ID NO: 2616)



TCTGCTCCCTCTGTGTCCTCA



(SEQ ID NO: 2617)










Also provided are sequences comprising the antisense sequences set forth above that are not the full length mRNA for any of the genes listed in Table 1 and can be used as antisense sequences. Further provided are antisense sequences that overlap with the sequences set forth above and comprise a fragment of the above-mentioned sequences. As mentioned above, these antisense sequences are merely exemplary, as it is known to those of skill in the art that once a mRNA sequence is provided for example the mRNA sequences set forth in Table 1, it is routine to walk along the mRNA sequence to generate antisense sequences that decrease expression. Therefore, the methods of the present invention can utilize any antisense sequence that decreases the expression of a gene set forth in Table 1.


shRNA


shRNA (short hairpin RNA) is a DNA molecule that can be cloned into expression vectors to express siRNA (typically 19-29 nt RNA duplex) for RNAi interference studies. shRNA has the following structural features: a short nucleotide sequence ranging from about 19-29 nucleotides derived from the target gene, followed by a short spacer of about 4-15 nucleotides (i.e. loop) and about a 19-29 nucleotide sequence that is the reverse complement of the initial target sequence.


Morpholinos

Morpholinos are synthetic antisense oligos that can block access of other molecules to small (about 25 base) regions of ribonucleic acid (RNA). Morpholinos are often used to determine gene function using reverse genetics methods by blocking access to mRNA. Morpholinos, usually about 25 bases in length, bind to complementary sequences of RNA by standard nucleic acid base-pairing. Morpholinos do not degrade their target RNA molecules. Instead, Morpholinos act by “steric hindrance”, binding to a target sequence within an RNA and simply interfering with molecules which might otherwise interact with the RNA. Morpholinos have been used in mammals, ranging from mice to humans.


Bound to the 5′-untranslated region of messenger RNA (mRNA), Morpholinos can interfere with progression of the ribosomal initiation complex from the 5′ cap to the start codon. This prevents translation of the coding region of the targeted transcript (called “knocking down” gene expression). Morpholinos can also interfere with pre-mRNA processing steps, usually by preventing the splice-directing snRNP complexes from binding to their targets at the borders of introns on a strand of pre-RNA. Preventing U1 (at the donor site) or U2/U5 (at the polypyrimidine moiety & acceptor site) from binding can cause modified splicing, commonly leading to exclusions of exons from the mature mRNA. Targeting some splice targets results in intron inclusions, while activation of cryptic splice sites can lead to partial inclusions or exclusions. Targets of U11/U12 snRNPs can also be blocked. Splice modification can be conveniently assayed by reverse-transcriptase polymerase chain reaction (RT-PCR) and is seen as a band shift after gel electrophoresis of RT-PCR products. Methods of designing, making and utilizing morpholinos are disclosed in U.S. Pat. No. 6,867,349 which is incorporated herein by reference in its entirety.


Small Molecules

Any small molecule that inhibits activity of a gene or a gene product set forth in Table 1 can be utilized in the methods of the present invention to decrease infection. These molecules are available in the scientific literature, in the StarLite/CHEMBL database available from the European Bioinformatics Institute, in DrugBank (Wishart et al. Nucleic Acids Res. 2006 Jan. 1; 34 (Database issue):D668-72), package inserts, brochures, chemical suppliers (for example, Sigma, Tocris, Aurora Fine Chemicals, to name a few), or by any other means, such that one of skill in the art makes the association between a gene product of Table 1 and inhibition of this gene product by a molecule. Preferred small molecules are those small molecules that have IC50 values of less than about 1 mM, less than about 100 micromolar, less than about 75 micromolar, less than about 50 micromolar, less than about 25 micromolar, less than about 10 micromolar, less than about 5 micromolar or less than about 1 micromolar. The half maximal inhibitory concentration (IC50) is a measure of the effectiveness of a compound in inhibiting biological or biochemical function. This quantitative measure indicates how much of a particular compound or other substance (inhibitor) is needed to inhibit a given biological process (or component of a process, i.e. an enzyme, cell, cell receptor or microorganism) by half. In other words, it is the half maximal (50%) inhibitory concentration (IC) of a substance (50% IC, or IC50). It is commonly used as a measure of antagonist drug potency in pharmacological research. Sometimes, it is also converted to the plC50 scale (−log IC50), in which higher values indicate exponentially greater potency. According to the FDA, IC50 represents the concentration of a drug that is required for 50% inhibition in vitro. It is comparable to an EC50 for agonist drugs. EC50 also represents the plasma concentration required for obtaining 50% of a maximum effect in vivo.


The present invention also provides the synthesis of small molecules that inhibit activity of a gene product set forth in Table 1. The present invention describes gene products for which three-dimensional structures are well known and can be obtained from the RCSB Protein Databank http://www.rcsb.org/pdb/home/home.do or http://www.rcsb.org for available three-dimensional structures. The structures and coordinates provided under the unique RCSB identifiers are hereby incorporated in their entireties by this reference. All of the structural information about the gene products set forth herein, for example, crystal structures and their corresponding coordinates, are readily available to one of skill in the art from the references cited herein, from the RCSB Protein Databank or elsewhere in the scientific literature.


Crystal structures can also be generated. Alternatively, one of skill in the art can obtain crystal structures for proteins, or domains of proteins, that are homologous to the proteins set forth in Table 1 from the RCSB Protein Databank or elsewhere in the scientific literature for use in homology modeling studies.


Routine high throughput in silico or in vitro screening of compound libraries for the identification of small molecules is also provided by the present invention. Compound libraries are commercially available. With an available crystal structure, it is routine for one of skill in the art to screen a library in silico and identify compounds with desirable properties, for example, binding affinity. For example, one of skill in the art can utilize the crystal structure(s) of a protein in a computer program to identify compounds that bind to a site on the crystal structure with a desirable binding affinity. This can be performed in an analogous way for any protein set forth herein to identify compounds that bind with a desirable binding affinity. Numerous computer programs are available and suitable for rational drug design and the processes of computer modeling, model building, and computationally identifying, selecting and evaluating potential compounds. These include, for example, SYBYL (available from TRIPOS, St. Louis Mo.), DOCK (available from University of California, San Francisco), GRID (available form Oxford University, UK), MCSS (available from Molecular Simulations Inc., Burlington, Mass.), AUTODOCK (available from Oxford Molecular Group), FLEX X (available from TRIPOS, St. Louis Mo.), CAVEAT (available from University of California, Berkeley), HOOK (available from Molecular Simulations Inc., Burlington, Mass.), and 3-D database systems such as MACCS-3D (available from MDL Information Systems, San Leandro, Calif.), UNITY (available from TRIPOS, St. Louis Mo.), and CATALYST (available from Molecular Simulations Inc., Burlington, Mass.). Compounds can also be computationally modified using such software packages as LUDI (available from Biosym TechMA), and LEAPFROG (TRIPOS Associates, St. Louis, Mo.). These computer-modeling techniques can be performed on any suitable hardware including for example, workstations available from Silicon Graphics, Sun Microsystems, and the like. These techniques, methods, hardware and software packages are representative and are not intended to be comprehensive listing. Other modeling techniques known in the art can also be employed in accordance with this invention.


A filter can be applied to the results to yield one or more compounds with a binding affinity in a particular range, for example, and not to be limiting, from about 100 micromolar to about 100 nanomolar, from about 10 micromolar to about 10 nanomolar, from about 1 micromolar to about 1 nanomolar, or from about 0.5 micromolar to about 0.5 nanomolar. Another filter can provide compounds with a certain binding affinity and size, for example, less than 1000 daltons, less than 500 daltons, less than 400 daltons, less than 300 daltons, less than 200 daltons, less than 100 daltons or less than 50 daltons or any size in between. The ranges and properties can be modified depending on the protein being studied. The compounds identified via this screening method can be further studied in silico, in vitro or in vivo. For example, the compounds can be modified in silico and rescreened in silico to determine the effects of chemical modifications on binding affinity or other properties being assessed in silico. The compounds identified in silico can be synthesized for in vitro or in vivo analysis.


All of the screening leading up to in vivo testing can be done in silico or in combination with in vitro assays. The initial compounds identified in silico and the resulting modified compounds can be screened in vitro, for example, in cellular assays to determine the effect of the compound on the cellular host protein as well as in viral assays, to determine antiviral activity. IC50 values can be obtained from the cellular assays, which may or may not be similar to the concentration necessary to effect 50% inhibition of viral infection in a viral assay. However, although not required, it is desirable to have a compound that has an IC50 value of less than about 1 mM, less than about 100 micromolar, less than about 75 micromolar, less than about 50 micromolar, less than about 25 micromolar, less than about 10 micromolar, less than about 5 micromolar or less than about 1 micromolar. Similarly, although not required, it is desirable to have a compound that effects 50% inhibition of viral infection at a concentration of less than about 1 mM, less than about 100 micromolar, less than about 75 micromolar, less than about 50 micromolar, less than about 25 micromolar, less than about 10 micromolar, less than about 5 micromolar or less than about 1 micromolar or any concentration in between.


Further modifications of the compounds can be done after in vitro screening, either in silico or via chemical synthesis, for further evaluation, prior to additional in vitro screening or in vivo studies. It is understood that this process can be iterative, involving a combination of in silico and wet chemistry techniques, but routine in drug development.


Other filters can be applied to the in silico screening process, for example, a filter that takes ADMET (adsorption, distribution, metabolism, excretion, toxicity) properties into consideration can be applied. ADMET modeling can be used during compound optimization to define an acceptable property space that contains compounds likely to have the desired properties. These filters can be applied sequentially or simultaneously depending.


Libraries for virtual or in vitro screening are available for the skilled artisan, for example from ChemBridge Corporation (San Diego, Calif.), such as a GPCR library, a kinase targeted library (KINACore), or an ion channel library (Ion Channel Set), to name a few. Compound libraries can also be obtained from the National Institutes of Health. For example, the NIH Clinical Collection of compounds that have been used in clinical trials can also be screened. Biofocus DPI (Essex, United Kingdom) also maintains and designs compound libraries that can be purchased for screening. One of skill in the art can select a library based on the protein of interest. For example, a kinase library can be screened to identify a compound that binds to and/or modulates a kinase. Other libraries that target enzyme families, for example, ATPases, hydrolases, isomerases, polymerases, transferases, phosphatases, etc., can also be screened, depending on the type of enzyme.


Compound libraries can also be screened in order to identify a compound that disrupts or inhibits specific interactions. Co-immunoprecipiation experiments can be utilized. Similarly, FRET analysis can be utilized, to identify compounds that disrupt the interaction between a two proteins.


Additional inhibitors include compositions comprising carbon and hydrogen, and optionally comprising one or more of —S, —N, —O, —Cl, —Br, or —Fl, appropriately bonded as a structure, with a size of less than about 1000 daltons, less than about 500 daltons, less than about 300 daltons, less than about 200 daltons, or less than about 100 daltons, that fits into a binding pocket or an active site of a gene product set forth herein. In particular, inhibitors that have the properties described in Lipinsky's Rule of Five are included herein. Lipinski's rule of five states that a drug/inhibitor has a weight under 500 Daltons, a limited lipophilicity or octanol-water partition coefficient (expressed by Log P<5, with P=[drug]org./[drug]aq.), a maximum of 5 H-bond donors (expressed as the sum of OHs and NHs), and a maximum of 10 H-bond acceptors (expressed as the sum of oxygen and nitrogen atoms). Inhibitors that violate no more than one of the above-listed five rules are also included herein.


CLTC

The following compounds are provided as inhibitors of CLTC. More specifically, the present invention provides a method of decreasing infection by a pathogen, in a cell or a subject said method comprising administering to the cell or subject an effective amount of a compound having the structure of a formula set forth in U.S. Pat. No. 3,310,469. U.S. Pat. No. 3,310,469 is hereby incorporated in its entirety by this reference, for its disclosure of CLTC inhibitors. An example of a CLTC inhibitor is set forth in formula I. Compounds of formula I can be made as described in U.S. Pat. No. 3,310,469. The pathogen can be a bacterium. The pathogen can also be a virus. The virus can be a gastrointestinal virus, as described herein. The virus can be a respiratory virus as described herein.




embedded image


MTAP, AHR, GLUD1, PLCB3, CRYZ, EEF1A1, MYC, NQO1, and TUBD1,

The following compounds are provided as inhibitors of MTAP, AHR, GLUD1, PLCB3, CRYZ, EEF1A1, MYC, NQO1, and TUBD1. More specifically, the present invention provides a method of decreasing infection by a pathogen, in a cell or a subject said method comprising administering to the cell or subject an effective amount of a compound having the structure of a formula set forth below.




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Other methods of decreasing expression and/or activity include methods of interrupting or altering transcription of mRNA molecules by site-directed mutagenesis (including mutations caused by a transposon or an insertional vector). Chemical mutagenesis can also be performed in which a cell is contacted with a chemical (for example ENU) that mutagenizes nucleic acids by introducing mutations into a gene set forth in Table 1. Transcription of mRNA molecules can also be decreased by modulating a transcription factor that regulates expression of any of the genes set forth in Table 1. Radiation can also be utilized to effect mutagenesis.


The present invention also provides decreasing expression and/or activity of a gene or a gene product set forth in Table 1 via modulation of other genes and gene products in pathways associated with the targets set forth in Table 1. Pathways include, but are not limited to ubiquitination pathways, trafficking pathways, cell signaling pathways, apoptotic pathways, TNF receptor pathways, GPCR pathways etc. Thus, other genes either upstream or downstream of the genes set forth in Table 1 are also provided herein in Table 2 as targets for inhibition of infection. For example, a gene product that interacts with MTAP either upstream or downstream in the MTAP pathway is considered a target for therapy or prevention against intracellular pathogens. For example, this can be a transcription factor that regulates expression of MTAP or another protein that interacts, either directly (for example, via binding to MTAP) or indirectly with MTAP. Examples of genes and gene products that can be modulated in this pathway include, but are not limited to those found in Table 2. These examples are merely exemplary as this applies to all of the genes and gene products set forth in Table 1 and the cellular pathways they are involved in. One of skill in the art would understand that modulation, including downregulation, upregulation, inhibition or stimulation of genes and/or gene products associated with the host cellular targets set forth in Table 1 can result in inhibition of infection. Such modulation can be effected by contacting a cell with a chemical, a small or large molecule (organic or inorganic), a drug, a protein, a peptide, a cDNA, an antibody, an aptamer, a morpholino, a triple helix molecule, an siRNA, a shRNA, an miRNA, an antisense RNA, an LNA, or a ribozyme which can be obtained via the methods set forth above. A combination of a composition that decreases expression and/or activity CLTC, for example, a CLTC inhibitor described herein, and a composition that modulates expression and/or activity of a CLTC modulator such as HIP1, etc. is further provided. Such combinations can be utilized to effect inhibition of infection by two or more, three or more, four or more, five or more pathogens set forth herein. In particular, these combinations can be utilized to inhibit infection by two or more, three or more, four or more; or five or more viruses. More particularly, these combinations can be utilized to inhibit infection by two or more, three or more, four or more; or five or more respiratory viruses. More particularly, these combinations can be utilized to inhibit infection by two or more, three or more, four or more; or five or more respiratory viruses selected from the group consisting of influenza, rhinovirus, adenovirus, parainfluenza virus, pox virus, RSV and measles. These combinations can also be utilized to inhibit infection by two or more strains of a respiratory virus selected from the group consisting of influenza, rhinovirus, adenovirus, parainfluenza virus, poxvirus, RSV and measles.










TABLE 2





Gene
Interactors/ Modulators







MTAP
SRM; APRT; AHCY; CDKN2A; ENSG00000037757; CDKN2B; DMBT1; AMD1;



IFNB1; HPRT1


AHR
AIP; ARNT; HSP90AA1; ESR1; RB1; CYP1A1; CYP1A2; CYP1B1; PTGES3;



CYP3A5


AK5
ADK; ADSL; AMPD3; AMPD1; APRT; ENTPD1; PNPT1; PDE11A; PKM2; NME4


AMOTL2
MCC; SERPINB3; RHOB; MPP1; GNG11; RBL2


ANXA4
GP2; SEC24D; SEC24B; SEC24A; SEC24C; STAM; SEC13; SEC23A; SFTPA2;



SFTPA1


ARL6IP5
SLC1A1; ARL6IP1; TBP; CCRS; HSPA4; GPS1; SLC1A2; C1orf122; RABAC1;



NIPSNAP3A


ARSA
GALC; GLA; UGT8; GLB1; ENSG00000206270; NEU4; NEU3; NEU2; GAL3ST1;



PSAP


ATOH8
RNF135; CTTNBP2; C20orf42; GBP1; IQGAP1; AKR1C3; CHI3L1; BMP6;



L1CAM; SMAD7


ATP6V1A
ATP6V1D; ATP6V0C; ATP6V0A4; ATP6V1B1; ATP6V1B2; ATP6V1F;



ATP6V0A1; ATP6V1H; ATP6V0D1; ATP6V1G1


BPNT1
PAPSS1; PAPSS2; GIYD1; SULT1A2; SULT1A1; CHST13; CHST11; CHST12;



SULT1E1; SULT2A1


C11ORF54
KIAA0423; DNAPTP6; HDHD2; TTLL12; TRIP13


C17ORF75
CHFR; LATS2; CDC25A; PIK3CA; RPS6KB1; ERBB3; FRAP1


C1ORF116
HOMER1; MIST; ACSM3; SLA; MGAM


CBLN2
CBLN3


CDH9
CDH10; CDH6; CDH15; CDH18; CTNNA1; CDH1; CTNND1; JUP; CDH8; CDH7


CEP152
TUBG2; XTP3TPATP1; CENPJ; ENSG00000103540; CEP78; MAPRE1;



CDK5RAP2; CEP110; CDC2; ODF2


CKAP2L
IL1RN; IL1A


CLTC
HIP1; CLINT1; ARF6; GGA2; CLTA; CLTB; AMPH; EPN1; OCRL; TFAP2A


CLUL1
MRLC3; EMILIN2; MYOM1; LPIN2; ZFP161; DLGAP1


CNTN5
PRL; IL2; PRLR; GHR; JAK2; C6orf66; CSH2; PTP4A3; CSN2; IRF1


CRYZ
NFE2L2; DECR1; JUN; CRYAA; CREB1; FOS; PPARA; NOS2A; NFKBIA;



ENSG00000206328


DAB2
MYO6; LRP2; GRB2; SMAD3; SMAD2; TGFBR1; TGFBR2; DAB2IP; CLTC;



SRC


DARS2
ADSS; CAD; ASNS; ADSSL1; ASPA; ASS1; GOT1; GAD1; GAD2; ACY3


DDIT4
TP53; TSC2; ATF4; INS; IGF1; RPS6KB1; FRAP1; RHEB; FKBP8; CRTC1


DDX42
SF3A2; SF3B1; DHX8; DDIT3; ASCC3L1; ETV1; SF3A1; POLD3; GSPT1; IFRD1


DNAH2
DNAH8; DNAH11; DNAH7; DNALI1; DNAH1; DNAH17; DNAH9; DNAH3;



DNAL4


DUSP5
MAPK3; MAPK1; TP53; MAPK8; MAPK9; ATF2; MAPK10; MAPK14; VEGFA;



MAPK13


EBNA1BP2
FGF3; CCNE1; BXDC1; E2F1; ENSG00000150991; GTPBP4; RRS1; CDK105;



ZCCHC8; TTC9C


EEF1A1
EEF1B2; RPS2; RPS7; TSFM; ZNF259; RPL7; RPL3; EEF1G; RPL5; RPL11


ENO1
PLG; TPI1; PKM2; PKLR; PGAM1; AK2; BPGM; ALDOA; PGAM2; ALDOC


ERGIC1
LMAN1; SURF4; ERGIC3; COPA; SURF1; EEF1A1


FAM35A
ENSG00000189014; ENSG00000122376


FKBP2
C1QC; ARFGEF2; SERPINB1; EPB41L2; EPB41; CDKN2A; PPIB; PPIA; MEN1;



UQCRH


FNDC3A
ITGA4; FN1; ITGA3; ITGAV; ITGA5; ITGB1; CNNM1; ACD; SDC1; CD44


FNDC3B
DYNC1LI1; TIMP4; CAPN2; GLS2; MGMT; NDP


GDF15
TP53; HAMP; ATF3; HLA-B; FOS; CD44; CDC25C; TLR9; CTSK; CA2


GEN1
TERF2; DNA2L; TNFRSF17; NCOA3; POLL


GLIS2
CTBP1; HNF4A; U2AF2; GPSM2; XAB2; CTNNB1; RBM9; CPSF1; SPECC1L;



WNK1


GLUD1
GLUL; CPS1; IDH2; GGT1; ASNS; GOT2; ADC; GAD2; ALDH18A1; EARS2


GLYCTK
HYI; KHK; PKM2; HIBADH; PKLR; ALDOB; ALDOA; ALDOC; PGP; SHMT2


GTF2H5
ERCC3; ERCC2; NR1H2; PMPCB; ENSG00000206370; XPA; GTF2H1; ERCC5;



CDK7; EXOSC7


HAVCR1
LGALS9; LCN2; TGM4; PHF11; ADAM33; ALPK2; CD68; CST3; NPSR1;



SEMA4A


HLA-DMA
ENSG00000206294; HLA-DMB; CIITA


HNRNPH3
ENSG00000211604; PSMD12; TRAF1; ATP5C1; ZMYM2; PCMT1; GABRP;



COX7C; HNRPM; SFRS2


HNRNPK
HNRPL; SRC; VAV1; KHDRBS1; PTBP1; PRMT1; RBMX; YBX1; SFRS3;



HNRPA1


IARS2
BCAT2; BCAT1; IARS; QRSL1; TAF1A; ESRRG; TP53BP2; MARS2; MARK1;



DCTD


IL18
CASP1; IFNG; IL18RAP; IL18R1; NOS2A; IL13; IL1B; IL1A; ENSG00000137496;



NOD2


IL6ST
IL6; LIF; PTPN11; JAK1; OSM; SOCS3; STAT3; IL11; STAT1; IL6R


ITGB1
ITGA2; ITGA3; ITGA5; PELO; ITGA6; ITGA4; ITGAV; ITGA9; ITGA11; ITGA8


KAZALD1
TMEM30B; WNT10A; DSPP; MAPK13; MMP20; WNT10B; DUSP6; ENPP1;



LEF1; DKK1


KCTD1
SLCO6A1; TFAP2A; HDAC9


KIAA1199
CRYM; COL9A3; KIAA1024; TBC1D2B; C15orf5; DNAJA4; IDH3A; WDR61;



TMED3; CIB2


KYNU
HAAO; KMO; TDO2; AFMID; INDO; ACMSD; AADAT; METTL6; PRMT8;



LCMT1


LMX1B
NPHS2; NPHS1; COL4A4; WNT7A; NKX2-2; GPM6A; COL4A3; FOXA2; SOX3;



FOXA1


LNX2
NUMB; LNX1; NUMBL; CXADR; TRAM2; CNTN1; TNFRSF12A; CNTN3;



RUNX2; PDZRN4


MALAT1
ATAD2; USP33; TFEB; SPON2; C1QB; IMPDH2; ACTN1; GBP1; NFIL3; EDIL3


MAPK1IP1L
AMH; CDK2AP2; ENOSF1; C1orf94; AHRR; SF3A2; STAP2


MAP4
MARK1; MAP7; EML1; MAP2; MAP1B; MAPT; TP53; MAP1A; KIFC1;



RABGAP1


MGAT3
MGAT4B; CDH1; MGAT4A; CTNNB1; CTNNA1; MGAT2; FUT8; CTNND1;



ANXA5; MGAT5


MICALCL
MAPK1; RAB1B


MLF1IP
CENPM; CENPO; CENPN; CENPI; CENPP; CENPQ; CENPT; CENPC1; CENPH;



MLF1


MLL5
C11orf30; SET; SETD7; P8; EXOSC7; CCNA2; NUP98; HOXA10; SMARCA4;



LBX1


MRPS18B
RB1; C6orf134; VARS2; KIAA1949; PSORS1C1; MRPS18A; PPP1R10; MDC1;



IER3; SP4


MTHFD1
SHMT1; MTR; MTHFR; TYMS; DHFR; ALDH1L1; MTHFD2L; MTHFS; FTCD;



SHMT2


MTMR11
NOC2L; SOCS3; BCL2; PPARG; RABEP2; TRA@; RASL12; IFNA1; IFNA17;



IFNA5


MYC
MAX; ZBTB17; TRRAP; BRCA1; RB1; CDKN2A; SMAD3; E2F1; E2F3; SP1


NAT13
RC3H1; HCN4; GJA5; USH1G; HCN1; GJA1; GSTZ1; HCN2; GJA7; CACNA1G


NDUFB3
NDUFA2; NDUFB5; NDUFS1; NDUFV1; NDUFAB1; CYCS; NDUFB1; NDUFA6;



NDUFS2; NDUFA1


NDUFS5
NDUFA2; NDUFS1; NDUFAB1; NDUFA7; NDUFV2; NDUFA5; NDUFS6;



NDUFS7; NDUFS2; NDUFS4


NFYA
NFYB; NFYC; ZHX1; TP53; RFXANK; ATF6; RFX5; ZHX3; DDIT3; OGG1


NPEPPS
KIAA1267; H3F3B; ITGB3; MAPT; TAF15; ELOVL2; PGPEP1; LRP8; VLDLR;



ATP6V0A1


NQO1
KEAP1; TP53; ODC1; CYP1A1; VKORC1; NFE2L2; GGCX; WWOX; SLC25A21;



MPO


NUAK2
ENSG00000150991; MAT1A; CAB39; ATIC; HSP90AA1; CDC37; NFKB2;



SMAD2; SMAD4; ACVR1


PAX8
TITF1; PPARG; TG; SLC5A5; WBP2; TSHR; FOXE1; FOXI1; LGALS3; RXR


PBLD
TMEM9; PDIA6; TTC5; STRAP; HMGCS1; GCH1; PNPO; GPAA1; CRYM;



ACSL4


PCBP2
SFRS3; HNRPL; PTBP1; HNRPD; HNRPK; YBX1; SFRS7; FUS; SFRS2; SRRM1


PDCD10
CCM2; STK25; KRIT1; STK24; PTPN13; ITGB1BP1; OSM; MYC; TEK; GLM


PKP1
DSP; DSG1; DSC1; JUP; KRT16; CDSN; A4GALT; VIM; DSG2; PLEKHG1


PLCB3
SLC9A3R2; GNAQ; CAMK2G; CAMK2B; ITPR1; EDN1; PIK3CB; PLCG1;



PIP5K1C; PLCD1


POLH
PCNA; REV1; RAD18; XPA; UBB; FUS; RAD51; OGG1; ERCC5; MSH2


PPP1R10
PPP1CA; C6orf134; VARS2; KIAA1949; MRPS18B; PSORS1C1; PPP1R7;



PPP1R8; KRT82; PPP1R1B


PTRH2
AES; PTH; PTHR2; PTHLH; PTHR1; ENSG00000142538; PTRH1; BCL2; FN1;



CHP


PXN
PTK2; GIT1; CRK; CSK; PTPN11; PTK2B; ILK; ITGB1; ITGA4; VCL


QARS
RARS; JTV1; MARS; KARS; LARS; IARS; SCYE1; EPRS; GMPS; GLUL


RAD51L1
RAD51C; HMGA2; RAD51L3; EVL; TP53; H1F0; TFAP2A; SLC20A1; XRCC2;



RAD1


RBMX
HNRPK; HTRA2; SF3A2; HNRPU; HNRPUL1; HNRPA1; PABPN1; NCBP1;



SFRS9; SF3B4


RMI1
TOP3A; C16orf75; BLM; WRN; RPA1; RPA2; C1orf173; C9orf64; GKAP1; TAF5L


RPL27A
RPL5; RPS29; RPS9; RPS10; RPL28; RPL21; RPL4; RPS3; RPL8; RPL11


RPL3
RPL4; RPL8; RPS3; RPL5; RPL11; RPL19; RPL17; RPL23; RPS11; RPL30


RPL34
RPL6; RPL4; RPL21; RPS29; RPS7; RPS23; RPS25; RPL30; RPL37; RPL35A


RPL37A
RPL35A; RPL27A; RPL37; RPL30; RPS3; RPL8; RPL4; RPS8; RPLP0; RPS19


RPL4
RPS3; RPL3; RPL5; RPL11; RPL8; RPL6; RPL3L; RPL19; RPL18; RPL17


RPL7A
RPL30; RPL4; RPL10A; RPL35A; RPS14; RPLP0; RPS23; RPS3; RPL6; RPLP2


RPS18
RPS14; POLR2C; MRPS11; MRPL17; RPS9; ENSG00000187928; RPL23;



ENSG00000172887; RPS3; POLR2L


RPSA
RPS21; LAMA2; RPS13; RPS29; RPS2; CBX5; RPS15; ENSG00000196084;



RPS14; RPS3


SDF2
CXCL12; SNAI2; GRASP; NFE2L2; DNAJB1; KDELR1; DNAJB11; IL9R;



MYSM1; PDIA4


SEMA3C
NRP1; NRP2; GATA6; SEMA3E; SEMA3A; SEMA3B; SEMA3G; SEMA3D;



SEMA3F; EPHA5


SERAC1
ASB9; TNFRSF19L; SYNJ2; PSMD1; HD; TERT


SERPINI1
PLAT; PLG; TTPA; PLAU; PDCD10; SLURP1; OTOL1; RAB33A; DEFB126;



COL15A1


SF3B4
SF3B2; SF3A2; SF3B1; SF3B5; P14; PHF5A; CD2BP2; CSTF1; SF3A3; THOC4


SFRS3
PCBP2; SFRS12; NXF1; HNRPK; SF3B2; THOC4; SF3A2; YBX1; HNRPL; CPSF3


SFXN1
SLC4A7; CDH17; SLC13A2; SLC19A1; CYBRD1; SLC7A9; SLC28A2; TFR2;



SLC22A5; SLC2A5


SKIL
SMAD3; SMAD2; SMAD4; NCOR1; RNF111; SMAD7; CREBBP; EP300;



HDAC1; CDH1


SLC25A25
CASP3; HINT2; PIK4CB; FREQ; NKX2-5; NBL1


SLC38A2
IGF1; MAPK8; SLC1A5; SLC7A6; SLC7A5; ATF4; SLC6A6; SLC43A2; SLC3A2;



SLC1A4


SLC39A14
DAPK3; SLC39A1; SLC30A10; SLC30A2; ZBTB11; LRRC28; SLC11A2;



SLC30A4; BTBD11; MS4A8B


SMC6
SMC5; CHEK2; RAD52; NSMCE2; MUS81; NOL6; ALK; DYM; DDX1; TSNAX


SPATA19
SH3GLB2; GOPC; LRP8; GPX4; HLA-A; CTLA4


SRP54
SRP19; SRP9; SRP14; MRPL27; SRPR; SRP72; SRP68; UCN2; RPS19; RPL23A


ST6GAL2
TMEM187; FLJ20019; FAM105A; FUT11; TMEM59L; SLC25A38; PIP5K1B;



EPHA6; HS3ST2; TMEM66


SUPT6H
SUPT5H; IWS1; SUPT16H; SMARCA1; SSRP1; POLE; SIRPA; SMARCC1; RTF1;



GTF2E1


TAF1
TBP; TAF7; TBN; TAF11; TAF6; TAF12; TAF2; TAF9; GTF2F1; TAF5


TBC1D5
MOCS3; SNX2; VPS26A; RUNX3; VPS35; LZTS1; SNX1; IGF2R; MORF4L1;



SERPINE2


TEX2
PDZD8; THOC3; SLC25A11; GIT1; PPARBP; ELAVL2


TMEM49
BECN1; TP53INP2; HEATR6; CHERP; APPBP2; TP53INP1; ARID5B; TUBD1;



CEACAM5; NFIB


TNPO1
HNRPA1; RAN; HNRPM; NUP98; RPL23A; HNRPD; NUP153; CCR2; RGPD6;



ELAVL1


TRAF7
MAP3K3; UBE2L3; FBXO25; FBXL5; FBXO7; FBXO4; ASMT; ARIH1; SUGT1;



FBXO10


TRIM66
TRIM28; TRIM33; TRIM24; CBX1; TRIM17; CBX3; TRIM45; CBX5


TSGA13
KLHDC10; TMEM209; TSGA14; CPA5; CPA4; ZC3HC1; CPA2; UBE2H; MEST;



CPA1


TUBD1
PACRG; HEATR6; YPEL2; PPM1E; APPBP2; TLK2; TEX14; STRBP; RSHL1;



SPAG4


TYW3
TYW1; TRMT12; LCMT2; AMMECR1; RPS19; PDCD5; ITGB4BP;



ENSG00000188463; PSMA3


UBA52
EGFR; TRAF6; TP53; MDM2; HIF1A; CBL; UBE2D1; PSMD4; IKBKG; UBE2N


USP10
G3BP1; SNX3; SCNN1A; IL17F


WDR51B
CUL4B


WDR82
SETD1A; CXXC1; SETD1B; RBBP5; ASH2L; SET; NR5A2; HCFC1; PHF7;



DPY30


WIPF2
WASL; WAS; NCK2; DNMBP; PDGFRB; PDGFRA; WASF1; BAZ1B;



SMARCA5; PFN1


ZHX2
AFP; GPC3; NFYA; RCBTB2; TSPYL1; ARID1B; ZHX3; ZHX1; SH3BGRL2;



CDC25C


ZMYND8
FHOD1; PRKCB1; WBP2; PTPRM; CEBPZ; SMARCA4; E2F6; SLC20A1; SPON1;



RECQL


ZNF143
POLR3F; POLR2K; POLR3D; POLR3E; POLR3B; SNAPC3; POLR3A; POU2F1;



POLR3H; POLR2L


ZWILCH
ZW10; KNTC1; ZWINT; BUB1B; MIS12; BUB3; BUB1; CDC20; NDC80; XPO1









Screening Methods

The present invention provides a method of identifying a compound that binds to a gene product set forth in Table 1 and can decrease infection of a cell by a pathogen comprising: a) contacting a compound with a gene product set forth in Table 1; b) detecting binding of the compound to the gene product; and c) associating the binding with a decrease in infection by the pathogen. This method can further comprise optimizing a compound that binds the gene product in an assay, for example, a cell based assay or an in vivo assay, that determines the functional ability to decrease infection. The binding assay can be a cellular assay or a non-cellular assay in which the gene product and the compound are brought into contact, for example, via immobilization of the gene product on a column, and subsequently contacting the immobilized gene product with the compound, or vice versa. Standard yeast two hybrid screens are also suitable for identifying a protein-protein interaction between a gene product set forth herein and another protein.


The present invention provides a method of identifying an agent that decreases infection of a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1; and b) detecting the level and/or activity of the gene product produced by the cellular gene, a decrease or elimination of the gene product and/or gene product activity indicating an agent with antipathogenic activity.


Also provided is a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1; b) contacting the cell with a pathogen; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.


Also provided is a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1, a decrease or elimination of the gene product and/or gene product activity indicating an agent with antipathogenic activity.


In the methods of the present invention, if the agent has previously been identified as an agent that decreases or inhibits the level and/or activity of a gene product set forth in Table 1, either via information in the literature or from in vitro or in vivo results, this can indicate a decrease in infection. A decrease in infection as compared to infection in a cell that was not contacted with the agent known to decrease or inhibit the level and/or activity of the gene product can be sufficient to identify the agent as an agent that decreases or inhibits infection.


The methods described herein can be utilized to identify any agent with an activity that decreases infection, prevents infection, or promotes cellular survival after infection with a pathogen(s). Therefore, the cell can be contacted with a pathogen before, or after being contacted with the agent. The cell can also be contacted concurrently with the pathogen and the agent. The agents identified utilizing these methods can be used to inhibit infection in cells either in vitro, in vivo, or ex vivo.


The present invention also provides a method of identifying a compound that binds to a gene product set forth in Table 1 and can decrease infection by three or more pathogens comprising: a) contacting a compound with a gene product set forth in Table 1; b) detecting binding of the compound to the gene product; and c) associating binding with a decrease in infection by three or more pathogens. This method can further comprise optimizing a compound that binds the gene product in an assay that determines the functional ability to decrease infection by three or more pathogens. This method can be cell based or an in vivo assay. The three or more pathogens can be any three or more pathogens set forth herein. For example, the three or more pathogens can be respiratory pathogens selected from the group consisting of picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses or adenoviruses. In another example, the three or more pathogens can be gastrointestinal pathogens selected from filoviruses, flaviviruses, calciviruses and reoviruses. The three or more pathogens can also be a combination of respiratory and gastrointestinal viruses. In another example, the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus. The cell population used in the method can be the same cell population for each pathogen or can be different cell populations. Typically, the agent would be administered to a different cell population for each pathogen assayed. For example, and not to be limiting, if the pathogens are viruses, a cell population is contacted with the agent and a first virus, another cell population is contacted with the agent and second virus, a third cell population is contacted with the agent and a third virus etc. in order to determine whether the agent inhibits infection by three or more viruses. Since the cell type will vary depending on whether or not a given virus can infect the cell, one of skill in the art would know how to pair the cell type with the virus in order to perform the assay.


This method can further comprise measuring the level of expression and/or activity of the gene product set forth in Table 1. This method can further comprise associating the level of infection with the level of expression and/or activity a gene product set forth in Table 1. In the screening methods disclosed herein, the level of infection can be measured, for example, by measuring viral replication.


In the methods of the present invention, if the agent has previously been identified as an agent that decreases or inhibits the level and/or activity of a gene product set forth in Table 1, this can indicate a decrease in infection. A decrease in infection as compared to infection in a cell that was not contacted with the agent known to decrease or inhibit the level and/or activity of the gene product can be sufficient to identify the agent as an agent that decreases or inhibits infection.


The methods described above can be utilized to identify any agent with an activity that decreases infection, prevents infection or promotes cellular survival after infection with a pathogen(s). Therefore, the cell can be contacted with a pathogen before, or after being contacted with the agent. The cell can also be contacted concurrently with the pathogen and the agent. The agents identified utilizing these methods can be used to inhibit infection in cells either in vitro, ex vivo or in vivo.


In the methods of the present invention any cell that can be infected with a pathogen can be utilized. The cell can be prokaryotic or eukaryotic, such as a cell from an insect, fish, crustacean, mammal, bird, reptile, yeast or a bacterium, such as E. coli. The cell can be part of an organism, or part of a cell culture, such as a culture of mammalian cells or a bacterial culture. The cell can also be in a nonhuman subject thus providing in vivo screening of agents that decrease infection by a pathogen. Cells susceptible to infection are well known and can be selected based on the pathogen of interest.


The test agents or compounds used in the methods described herein can be, but are not limited to, chemicals, FDA approved drugs, clinical compounds, European approved drugs, Japanese approved drugs, small molecules, inorganic molecules, organic molecules, drugs, proteins, cDNAs, large molecules, antibodies, aptamers, morpholinos, triple helix molecule, peptides, siRNAs, shRNAs, miRNAs, antisense RNAs, LNAs, ribozymes or any other compound. The compound can be random or from a library optimized to bind a gene product as set forth in Table 1. Drug libraries optimized for the proteins in the class of proteins provided herein can also be screened or tested for binding or activity. Compositions identified with the disclosed approaches can be used as lead compositions to identify other compositions having even greater antipathogenic activity. For example, chemical analogs of identified chemical entities, or variants, fragments or fusions of peptide agents, can be tested for their ability to decrease infection using the disclosed assays. Candidate agents can also be tested for safety in animals and then used for clinical trials in animals or humans.


In the methods described herein, once the cell containing a cellular gene encoding a gene product set forth in Table 1 has been contacted with an agent, the level of infection can be assessed by measuring an antigen or other product associated with a particular infection. For example, the level of viral infection can be measured by real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR) assay (See for example, Payungporn et al. “Single step multiplex real-time RT-PCR for H5N1 influenza A virus detection.” J Virol Methods. Sep. 22, 2005; Landolt et al. “Use of real-time reverse transcriptase polymerase chain reaction assay and cell culture methods for detection of swine influenza A viruses” Am J Vet Res. 2005 January; 66(1):119-24). If there is a decrease in infection then the composition is an effective agent that decreases infection. This decrease does not have to be complete as the decrease can be a 10%, 20%, 30%, 40%, 50%, 60%. 70%, 80%, 90%, 100% decrease or any percentage decrease in between.


In the methods set forth herein, the level of the gene product can be measured by any standard means, such as by detection with an antibody specific for the protein. The nucleic acids set forth herein and fragments thereof can be utilized as primers to amplify nucleic acid sequences, such as a gene transcript of a gene set forth in Table 1 by standard amplification techniques. For example, expression of a gene transcript can be quantified by real time PCR using RNA isolated from cells. A variety of PCR techniques are familiar to those skilled in the art. For a review of PCR technology, see White (1997) and the publication entitled “PCR Methods and Applications” (1991, Cold Spring Harbor Laboratory Press), which is incorporated herein by reference in its entirety for amplification methods. In each of these PCR procedures, PCR primers on either side of the nucleic acid sequences to be amplified are added to a suitably prepared nucleic acid sample along with dNTPs and a thermostable polymerase such as Taq polymerase, Pfu polymerase, or Vent polymerase. The nucleic acid in the sample is denatured and the PCR primers are specifically hybridized to complementary nucleic acid sequences in the sample. The hybridized primers are extended. Thereafter, another cycle of denaturation, hybridization, and extension is initiated. The cycles are repeated multiple times to produce an amplified fragment containing the nucleic acid sequence between the primer sites. PCR has further been described in several patents including U.S. Pat. Nos. 4,683,195, 4,683,202 and 4,965,188. Each of these publications is incorporated herein by reference in its entirety for PCR methods. One of skill in the art would know how to design and synthesize primers that amplify any of the nucleic acid sequences set forth herein or a fragment thereof.


A detectable label may be included in an amplification reaction. Suitable labels include fluorochromes, e.g. fluorescein isothiocyanate (FITC), rhodamine, Texas Red, phycoerythrin, allophycocyanin, 6-carboxyfluorescein (6-FAM), 2′,7′-dimethoxy-4′,5′-dichloro-6-carboxyfluorescein (JOE),6-carboxy-X-rhodamine (ROX), 6-carboxy-2′,4′,7′,4,7-hexachlorofluorescein (HEX), 5-carboxyfluorescein (5-FAM) or N,N,N′,N′-tetramethyl-6-carboxyrhodamine (TAMRA), radioactive labels, e.g., 32P, 35S, 3H; etc. The label may be a two stage system, where the amplified DNA is conjugated to biotin, haptens, etc. having a high affinity binding partner, e.g. avidin, specific antibodies, etc., where the binding partner is conjugated to a detectable label. The label may be conjugated to one or both of the primers. Alternatively, the pool of nucleotides used in the amplification is labeled, so as to incorporate the label into the amplification product.


The sample nucleic acid, e.g. amplified fragment, can be analyzed by one of a number of methods known in the art. The nucleic acid can be sequenced by dideoxy or other methods. Hybridization with the sequence can also be used to determine its presence, by Southern blots, dot blots, etc.


In the methods of the present invention, the level of gene product can be compared to the level of the gene product in a control cell not contacted with the compound. The level of gene product can be compared to the level of the gene product in the same cell prior to addition of the compound. The activity or the level of gene product can be compared to the activity or the level of the gene product in the same cell prior to addition of the compound. The activity or level of the gene product can also be compared to the activity or the level of the gene product in a control cell contacted with a compound known to decrease the activity and/or the level of the gene product. Activity or function, can be measured by any standard means, for example, and not to be limiting, by enzymatic assays that measure the conversion of a substrate to a product, by signal transduction assays, or binding assays that measure the binding of a gene product set forth in Table 1 to another protein, for example.


Moreover, the regulatory region of a gene set forth in Table 1 can be functionally linked to a reporter gene and compounds can be screened for inhibition of reporter gene expression. Such regulatory regions can be isolated from genomic sequences and identified by any characteristics observed that are characteristic for regulatory regions of the species and by their relation to the start codon for the coding region of the gene. As used herein, a reporter gene encodes a reporter protein. A reporter protein is any protein that can be specifically detected when expressed. Reporter proteins are useful for detecting or quantitating expression from expression sequences. Many reporter proteins are known to one of skill in the art. These include, but are not limited to, β-galactosidase, luciferase, and alkaline phosphatase that produce specific detectable products. Fluorescent reporter proteins can also be used, such as green fluorescent protein (GFP), cyan fluorescent protein (CFP), red fluorescent protein (RFP) and yellow fluorescent protein (YFP).


Viral infection can also be measured via cell based assays. Briefly, by way of example, cells (20,000 to 2,500,000) are infected with the desired pathogen, and the incubation continued for 3-7 days. The antiviral agent can be applied to the cells before, during, or after infection with the pathogen. The amount of virus and agent administered can be determined by skilled practitioners. In some examples, several different doses of the potential therapeutic agent can be administered, to identify optimal dose ranges. Following transfection, assays are conducted to determine the resistance of the cells to infection by various agents. For example, if analyzing viral infection, the presence of a viral antigen can be determined by using antibody specific for the viral protein then detecting the antibody. In one example, the antibody that specifically binds to the viral protein is labeled, for example with a detectable marker such as a fluorophore. In another example, the antibody is detected by using a secondary antibody containing a label. The presence of bound antibody is then detected, for example using microscopy, flow cytometry and ELISA. In any of the methods set forth herein, the amount of viral inhibition can be compared to the amount of viral inhibition in a control cell contacted with an agent that is known to decrease viral inhibition. For example, and not to be limiting, for influenza, the amount of viral inhibition can be compared to the amount of viral inhibition in a control cell contacted with Tamiflu, amantadine, ribavirin, Relenza etc. Similar approaches can be utilized with any other virus or pathogen for which there is a known inhibitor of viral infection that can be utilized as a positive control. Similar methods can be used to monitor bacterial, protozoal, or fungal infection (except that the antibody would recognize a bacterial, protozoal, or fungal protein, respectively).


For example, if analyzing viral infection, the presence of a viral antigen can be determined by using antibody specific for the viral protein then detecting the antibody. In one example, the antibody that specifically binds to the viral protein is labeled, for example with a detectable marker such as a fluorophore. In another example, the antibody is detected by using a secondary antibody containing a label. The presence of bound antibody is then detected, for example using microscopy, flow cytometry and ELISA. Similar methods can be used to monitor bacterial, protozoal, or fungal infection (except that the antibody would recognize a bacterial, protozoal, or fungal protein, respectively).


Alternatively, or in addition, the ability of the cells to survive viral infection is determined, for example, by performing a cell viability assay, such as trypan blue exclusion. Plaque assays can be utilized as well.


The amount of protein in a cell, can be determined by methods standard in the art for quantitating proteins in a cell, such as Western blotting, ELISA, ELISPOT, immunoprecipitation, immunofluorescence (e.g., FACS), immunohistochemistry, immunocytochemistry, etc., as well as any other method now known or later developed for quantitating protein in or produced by a cell.


The amount of a nucleic acid in a cell can be determined by methods standard in the art for quantitating nucleic acid in a cell, such as in situ hybridization, quantitative PCR, RT-PCR, Taqman assay, Northern blotting, ELISPOT, dot blotting, etc., as well as any other method now known or later developed for quantitating the amount of a nucleic acid in a cell.


Any of the screening methods set forth herein can optionally comprise the step of assessing toxicity of a composition via any of the toxicity measurement methods described herein, or via any of the toxicity measurement methods known to one of skill in the art, such as, for example, the CytoTox-Glo assay (see Niles, A. et al. (2007) Anal. Biochem. 366, 197-206) or the Cell-Titer-Glo assay from Promega.


The ability of an antiviral agent to prevent or decrease infection by a virus, for example, any of the viruses listed above, can be assessed in an animal model. Several animal models for viral infection are known in the art. For example, mouse HIV models are disclosed in Sutton et al. (Res. Initiat Treat. Action, 8:22-4, 2003) and Pincus et al. (AIDS Res. Hum. Retroviruses 19:901-8, 2003); guinea pig models for Ebola infection are disclosed in Parren et al. (J. Virol. 76:6408-12, 2002) and Xu et al. (Nat. Med. 4:37-42, 1998); cynomolgus monkey (Macaca fascicularis) models for influenza infection are disclosed in Kuiken et al. (Vet. Pathol. 40:304-10, 2003); mouse models for herpes are disclosed in Wu et al. (Cell Host Microbe 22:5(1):84-94. 2009); pox models are disclosed in Smee et al. (Nucleosides Nucleotides Nucleic Acids 23(1-2):375-83, 2004) and in Bray et al. (J. Infect. Dis. 181(1):10-19); and Franciscella tularensis models are disclosed in Klimpel et al. (Vaccine 26(52): 6874-82, 2008).


Other animal models for influenza infection are also available. These include, but are not limited to, a cotton rat model disclosed by Ottolini et al. (J. Gen. Virol., 86(Pt 10): 2823-30, 2005), as well as ferret and mouse models disclosed by Maines et al. (J. Virol. 79(18):11788-11800, 2005). One of skill in the art would know how to select an animal model for assessing the in vivo activity of an agent for its ability to decrease infection by viruses, bacteria, fungi and parasites.


Such animal models can also be used to test agents for an ability to ameliorate symptoms associated with viral infection. In addition, such animal models can be used to determine the LD50 and the ED50 in animal subjects, and such data can be used to determine the in vivo efficacy of potential agents. LD50 is an index of toxicity (lethal dose 50%), the amount of the substance that kills 50% of the test population of experimental animals when administered as a single dose. ED50 is the dose of a drug that is pharmacologically effective for 50% of the population exposed to the drug or a 50% response in a biological system that is exposed to the drug. Animal models can also be used to assess antibacterial, antifungal and antiparasitic agents.


Animals of any species, including, but not limited to, birds, ferrets, cats, mice, rats, rabbits, fish (for example, zebrafish) guinea pigs, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, and chimpanzees, can be used to generate an animal model of viral infection, bacterial infection, fungal infection or parasitic infection if needed.


For example, for a model of viral infection, the appropriate animal is inoculated with the desired virus, in the presence or absence of the antiviral agent. The amount of virus and agent administered can be determined by skilled practitioners. In some examples, several different doses of the potential therapeutic agent (for example, an antiviral agent) can be administered to different test subjects, to identify optimal dose ranges. The therapeutic agent can be administered before, during, or after infection with the virus. Subsequent to the treatment, animals are observed for the development of the appropriate viral infection and symptoms associated therewith. A decrease in the development of the appropriate viral infection, or symptoms associated therewith, in the presence of the agent provides evidence that the agent is a therapeutic agent that can be used to decrease or even inhibit viral infection in a subject. For example, a virus can be tested which is lethal to the animal and survival is assessed. In other examples, the weight of the animal or viral titer in the animal can be measured. Similar models and approaches can be used for bacterial, fungal and parasitic infections.


In the methods of the present invention, the level of infection can be associated with the level of gene expression and/or activity, such that a decrease or elimination of infection associated with a decrease or elimination of gene expression and/or activity indicates that the agent is effective against the pathogen. For example, the level of infection can be measured in a cell after administration of siRNA that is known to inhibit a gene product set forth in Table 1. If there is a decrease in infection then the siRNA is an effective agent that decreases infection. This decrease does not have to be complete as the decrease can be a 10%, 20%, 30%, 40%, 50%, 60%. 70%, 80%, 90%, 100% decrease or any percentage decrease in between. In the event that the compound is not known to decrease expression and/or activity of a gene product set forth in Table 1, the level of expression and/or activity of can be measured utilizing the methods set forth above and associated with the level of infection. By correlating a decrease in expression and/or activity with a decrease in infection, one of skill in the art can confirm that a decrease in infection is effected by a decrease in expression and/or activity of a gene or gene product set forth in Table 1. Similarly, the level of infection can be measured in a cell, utilizing the methods set forth above and known in the art, after administration of a chemical, small molecule, drug, protein, cDNA, antibody, aptamer, shRNA, miRNA, morpholino, antisense RNA, ribozyme or any other compound. If there is a decrease in infection, then the chemical, small molecule, drug, protein, cDNA, antibody, shRNA, miRNA, morpholino, antisense RNA, ribozyme or any other compound is an effective antpathogenic agent.


The present invention provides a method of identifying an agent that can decrease infection by two or more pathogens comprising: a) administering the agent to two or more cell populations containing a cellular gene encoding a gene product set forth in Table 1; b) contacting the two or more cell populations with a pathogen, wherein each population is contacted with a different pathogen; and c) determining the level of infection, a decrease or elimination of infection by two or more pathogens indicating that the agent is an agent that decreases infection by three or more pathogens.


The present invention provides a method of identifying an agent that can decrease infection by three or more pathogens comprising: a) administering the agent to three or more cell populations containing a cellular gene encoding a gene product set forth in Table 1; b) contacting the three or more cell populations with a pathogen, wherein each population is contacted with a different pathogen; and c) determining the level of infection, a decrease or elimination of infection by three or more pathogens indicating that the agent is an agent that decreases infection by three or more pathogens.


It is understood that two or more, also means three or more, four or more, five or more, six or more, seven or more, etc. Therefore, the screening methods set forth above can be utilized to identify agents that decrease infection by four or more, five or more, six or more, seven or more pathogens set forth herein.


More particularly, the two or more, three or more, four or more, five or more, six or more, or seven or more pathogens can be selected from the group consisting of Franscicella tularensis, a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus and an adenovirus. The two or more, three or more, four or more, five or more, six or more, or seven or more pathogens can also be selected from the group consisting of Franscicella tularensis, a filovirus, an adenovirus, a picornavirus, a calicivirus, a flavivirus and a reovirus. The two or more, three or more, four or more, five or more, six or more, or seven or more pathogens can also be selected from the group consisting of Franscicella tularensis, a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, an adenovirus, a filovirus, a picornavirus, a calicivirus, a flavivirus and a reovirus.


The two or more, three or more, four or more, five or more pathogens can also be selected from the group consisting of Franscicella tularensis, influenza, rhinovirus, parainfluenza virus, measles, a pox virus and RSV. The two or more, three or more, four or more, five or more, six or more, or seven or more pathogens can also be selected from the group consisting of Franscicella tularensis, a reovirus, an adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a Dengue fever virus, a West Nile virus, a yellow fever virus, a rotavirus and an enterovirus. The two or more, three or more, four or more, five or more, six or more, or seven or more pathogens can also be selected from the group consisting of Franscicella tularensis, HIV, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, a reovirus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus. The two or more, three or more, four or more, five or more, six or more, or seven or more pathogens can also be selected from the group consisting of Franscicella tularensis, influenza, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus. The two or more, three or more, four or more, five or more, six or more, or seven or more pathogens can also be selected from the group consisting of Franscicella tularensis, an HIV, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, tuberculosis, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


The cell population used in the assay can be the same cell population for each virus strain or can be different cell populations. Typically, the agent would be administered to a different cell population for each viral strain assayed. For example, and not to be limiting, a cell population is contacted with the agent and a first virus, another cell population is contacted with the agent and second virus, a third cell population is contacted with the agent and a third virus etc. in order to determine whether the agent inhibits infection by three or more pathogens. Since the cell type will vary depending on whether or not a given virus can infect the cell, one of skill in the art would know how to pair the cell type with the virus in order to perform the assay.


This method can further comprise measuring the level of expression and/or activity of a gene product set forth in Table 1. This method can further comprise associating the level of infection with the level of expression and/or activity of a gene product set forth in Table 1. In the screening methods disclosed herein, the level of infection can be measured, for example, by measuring viral load as described in the Examples. In any of the screening methods described throughout this application, one of skill in the art can compare the level of infection in a cell contacted with a test agent with a cell contacted with a compound that is known to decrease infection in a cell, for example, a compound that targets a viral protein, in order to compare the level of infection with a positive control.


Further provided by the present invention is a method of identifying an agent that can decrease infection by three or more pathogens comprising: a) administering the agent to three or more cell populations containing a cellular gene encoding a gene product set forth in Table 1; b) contacting the three or more cell populations with a pathogen, wherein each population is contacted with a different pathogen; and c) determining the level of expression and/or activity of the gene product, a decrease or elimination of gene product expression or activity in cells indicating that the agent is an agent that decreases infection by three or more pathogens.


In the methods of the present invention, if the compound has previously been identified as a compound that decreases or inhibits the level and/or activity of the gene product, for example, via the scientific literature, in vitro studies or in vivo studies, it is not necessary to associate a decrease in infection with the level/and or activity of the gene product. A decrease in infection as compared to infection in a cell that was not contacted with the agent known to decrease or inhibit the level and/or activity of the gene product is sufficient to identify the agent as an agent that decreases or inhibits infection.


The methods described above can be utilized to identify any compound with an activity that decreases infection, prevents infection or promotes cellular survival after infection with a pathogen(s). Therefore, the cell can be contacted with a bacterium or a virus before, or after being contacted with the agent. The cell can also be contacted concurrently with the bacterium or the virus and the agent. The compounds identified utilizing these methods can be used to inhibit infection in cells either in vitro, ex vivo or in vivo.


In the methods of the present invention any cell that can be infected with a bacterium or a virus can be utilized. The cell can be prokaryotic or eukaryotic, such as a cell from an insect, fish, crustacean, mammal, bird, reptile, yeast or a bacterium, such as E. coli. The cell can be part of an organism, or part of a cell culture, such as a culture of mammalian cells or a bacterial culture. The cell can also be in a nonhuman subject thus providing in vivo screening of agents that decrease infection by a pathogen. Cells susceptible to viral infection are well known and would be selected based on the pathogen of interest.


Compositions identified with the disclosed approaches can be used as lead compositions to identify other compositions having even greater antipathogenic activity. For example, chemical analogs of identified chemical entities, or variants, fragments or fusions of peptide agents, can be tested for their ability to decrease infection using the disclosed assays. Candidate agents can also be tested for safety in animals and then used for clinical trials in animals or humans.


It is understood that any of the screening methods described herein can be performed in any tissue culture dish, including but not limited to 6 well, 12 well, 24 well, 96 well or 384 well plates. The assays can also be automated by utilizing robotics and other instrumentation standard in the art of drug screening.


Arrays

The genes and nucleic acids of the invention can also be used in polynucleotide arrays. Polynucleotide arrays provide a high throughput technique that can assay a large number of polynucleotide sequences in a single sample. This technology can be used, for example, to identify samples with reduced expression of as compared to a control sample. This technology can also be utilized to determine the effects of reduced expression of a gene set forth in Table 1 on other genes. In this way, one of skill in the art can identify genes that are upregulated or downregulated upon reducing expression of a gene set forth in Table 1. Similarly, one of skill in the art can identify genes that are upregulated or downregulated upon increased expression of a gene set forth in Table 1. This allows identification of other genes that are upregulated or downregulated upon modulation of expression that can be targets for therapy, such as antiviral therapy, antibacterial therapy, antiparasitic therapy or antifungal therapy.


To create arrays, single-stranded polynucleotide probes can be spotted onto a substrate in a two-dimensional matrix or array. Each single-stranded polynucleotide probe can comprise at least 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, or 30 or more contiguous nucleotides selected from nucleotide sequences set forth under GenBank Accession Nos. herein and other nucleic acid sequences that would be selected by one of skill in the art depending on what genes, in addition to one ore more of the genes set forth in Table 1, 2, 3 or 4 are being analyzed.


The array can also be a microarray that includes probes to different polymorphic alleles of these genes. A polymorphism exists when two or more versions of a nucleic acid sequence exist within a population of subjects. For example, a polymorphic nucleic acid can be one where the most common allele has a frequency of 99% or less. Different alleles can be identified according to differences in nucleic acid sequences, and genetic variations occurring in more than 1% of a population (which is the commonly accepted frequency for defining polymorphism) are useful polymorphisms for certain applications. The allelic frequency (the proportion of all allele nucleic acids within a population that are of a specified type) can be determined by directly counting or estimating the number and type of alleles within a population. Polymorphisms and methods of determining allelic frequencies are discussed in Hartl, D. L. and Clark, A. G., Principles of Population Genetics, Third Edition (Sinauer Associates, Inc., Sunderland Mass., 1997), particularly in chapters 1 and 2.


These microarrays can be utilized to detect polymorphic alleles in samples from subjects. Such alleles may indicate that a subject is more susceptible to infection or less susceptible to infection. For example, microarrays can be utilized to detect polymorphic versions of genes set forth in Table 1 that result in decreased gene expression and/or decreased activity of the gene product to identify subjects that are less susceptible to viral infection. In addition, the existence of an allele associated with decreased expression in a healthy individual can be used to determine which genes are likely to have the least side effects if the gene product is inhibited or bound or may be selected for in commercial animals and bred into the population.


The substrate can be any substrate to which polynucleotide probes can be attached, including but not limited to glass, nitrocellulose, silicon, and nylon. Polynucleotide probes can be bound to the substrate by either covalent bonds or by non-specific interactions, such as hydrophobic interactions. Techniques for constructing arrays and methods of using these arrays are described in EP No. 0 799 897; PCT No. WO 97/29212; PCT No. WO 97/27317; EP No. 0 785 280; PCT No. WO 97/02357; U.S. Pat. Nos. 5,593,839; 5,578,832; EP No. 0 728 520; U.S. Pat. No. 5,599,695; EP No. 0 721 016; U.S. Pat. No. 5,556,752; PCT No. WO 95/22058; and U.S. Pat. No. 5,631,734. Commercially available polynucleotide arrays, such as Affymetrix GeneChip™ can also be used. Use of the GeneChip™ to detect gene expression is described, for example, in Lockhart et al., Nature Biotechnology 14:1675 (1996); Chee et al., Science 274:610 (1996); Hacia et al., Nature Genetics 14:441, 1996; and Kozal et al., Nature Medicine 2:753, 1996.


Methods of Making Compounds

The present invention provides a method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 1; c) contacting the cell with an infectious pathogen; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product.


Further provided is a method of making a compound that decreases infection in a cell by a pathogen, comprising: a) optimizing a compound to bind a gene product set forth in Table 1; b) administering the compound to a cell containing a cellular gene encoding the gene product; c) contacting the cell with an infectious pathogen; d) determining the level of infection, a decrease or elimination of infection indicating the making of a compound that decreases infection in a cell by a pathogen. This method can further synthesizing therapeutic quantities of the compound.


The present invention also provides a method of synthesizing a compound that binds to a gene product set forth in Table 1 and decreases infection by a pathogen comprising: a) contacting a library of compounds with a gene product set forth in Table 1; b) associating binding with a decrease in infection; and c) synthesizing derivatives of the compounds from the library that bind to the gene product.


The present invention also provides a business method to reduce the cost of drug discovery of drugs that can reduce infection by a pathogen comprising: screening, outside of the United States, for drugs that reduce infection by binding to or reducing the function of a gene product set forth in Table 1; and b) importing drugs that reduce infection into the United States. Also provided is a method of making drugs comprising directing the synthesis of drugs that reduce infection by binding to or reducing the function of a gene or gene product set forth in Table 1.


Pharmaceutical Compositions and Modes of Administration

The present invention provides a method of decreasing infection by a pathogen in a subject by decreasing the expression or activity of a gene or gene product set forth in Table 1, said method comprising administering to the subject an effective amount of a composition that decreases the expression or activity of a gene or a gene product set forth in Table 1. It is understood that in this method, the method is not limited to the decrease in expression and/or activity of one gene or gene product, as more than one gene or gene product, for example, two, three, four, five, six, etc. can be inhibited in order to inhibit infection by a pathogen.


The composition can comprise one or more of, a chemical, a compound, a small molecule, an inorganic molecule, an organic molecule, a drug, a protein, a cDNA, a peptide, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an miRNA, an antisense nucleic acid or a ribozyme that decreases the expression or activity of one or more of the genes or gene products of Table 1.


A composition can also be a mixture, cocktail or combination of two or more compositions, for example, two or more compositions selected from the group consisting of chemical, a compound, a small molecule, an inorganic molecule, an organic molecule, an aptamer, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an LNA, an antisense nucleic acid or a ribozyme. The two or more compositions can be the same or different types of compositions. For example, and not to be limiting two or more compositions can be an antisense and a small molecule; or two antisense molecules; or two small molecules; or an siRNA and small molecule, etc. It is understood that any combination of the types of compositions set forth herein can be utilized in the methods set forth herein.


Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by two or more respiratory viruses. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by three or more respiratory viruses. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by four or more respiratory viruses. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by five or more respiratory viruses. These can be selected from the group consisting of: a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus and an adenovirus. Since picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses and adenoviruses are families of viruses, two or more, three or more, four or more, or five or more respiratory viruses can be from the same or from different families. For example, and not to be limiting, the composition can inhibit infection by two or more orthomyxoviruses; two or more picornaviruses; an orthomyxovirus, an adenovirus, and a picornavirus; an orthomyxovirus, a paramyxovirus and an adenovirus; an orthomyxovirus, two picornaviruses and a paramyxovirus; three orthomyxoviruses, a picornavirus and an adenovirus, etc. More particularly, the composition can inhibit infection by two or more, three or more or four or more respiratory viruses selected from the group consisting of an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus and an RSV virus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by two or more gastrointestinal viruses. The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by three or more gastrointestinal viruses. The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by four or more gastrointestinal viruses. The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by five or more gastrointestinal viruses. These viruses can be selected from the group consisting of: a filovirus, a picornavirus, a calcivirus, a flavivirus or a reovirus. Since filoviruses, picornaviruses, calciviruses, flaviviruses and reoviruses are families of viruses, the composition can inhibit infection by two or more, three or more, four or more, or five or more gastrointestinal viruses from the same or from different families. More particularly, the composition can inhibit infection by two or more, three or more, four or more, or five or more gastrointestinal viruses selected from the group consisting of a reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a Dengue fever virus, a West Nile virus, a yellow fever virus, a rotavirus and an enterovirus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by one or more pathogens selected from the group consisting of: a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, an adenovirus, and inhibits infection by one or more pathogens selected from the group consisting of: a flavivirus, a filovirus, a calcivirus or a reovirus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by two or more pathogens selected from the group consisting of HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1 wherein the composition inhibits infection by two or more pathogens selected from the group consisting of: influenza, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by three or more pathogens. The three or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein. More particularly, the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by four or more pathogens. The four or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein. More particularly, the four or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by five or more pathogens. The five or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein. More particularly, the five or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by six or more pathogens. The six or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein. More particularly, the six or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits co-infection by HIV and one or more viruses, bacteria, parasites or fungi. For example, decreasing co-infection of HIV and any of the viruses, including for example any families, genus, species, or group of viruses. As a further example, co-infection of HIV and a respiratory virus is provided herein. Respiratory viruses include picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses, and adenoviruses. More specifically, the respiratory virus can be any strain of influenza, rhinovirus, adenovirus, parainfluenza virus or RSV. Also provided is decreasing co-infection of HIV and a gastrointestinal virus. Gastrointestinal viruses include picornaviruses, filoviruses, flaviviruses, calciviruses and reoviruses. More specifically, and not to be limiting, the gastrointestinal virus can be any strain of reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus. Further provided is a method of decreasing co-infection of HIV with a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus. More particularly, decreasing co-infection of HIV and a hepatitis virus, such as Hepatitis A, Hepatitis B or Hepatitis C is provided. Also provided is decreasing co-infection of HIV and a herpes virus, for example, HSV-1 or HSV-2. In addition decreasing co-infection of HIV and tuberculosis is also provided. Further provided is decreasing co-infection of HIV and CMV, as well as decreasing co-infection of HIV and HPV.


As described herein, the genes set forth in Tables 1 can be involved in the pathogenesis of two or more respiratory viruses. Therefore, the present invention provides methods of treating or preventing an unspecified respiratory infection in a subject by administering a composition that decreases activity or expression of a gene involved in the pathogenesis of two or more respiratory viruses. More particularly, the present invention provides a method of decreasing an unspecified respiratory infection in a subject comprising: a) diagnosing a subject with an unspecified respiratory infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by two or more respiratory viruses selected from the group consisting of picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses, or adenoviruses. As set forth above, in the methods of the present invention, the two or more respiratory viruses can be from the same family or from a different family of respiratory viruses. More specifically, the respiratory virus can be any strain of influenza, rhinovirus, adenovirus, parainfluenza virus or RSV. In this method, the composition can be a composition that inhibits infection by three or more, four or more, five or more; or six or more respiratory viruses selected from the group consisting of a picornaviruses, an orthomyxoviruses, paramyxoviruses, coronaviruses, or adenoviruses.


As described herein, the genes set forth in Tables 1 can be involved in the pathogenesis of two or more gastrointestinal viruses. Therefore, the present invention provides methods of treating or preventing an unspecified gastrointestinal infection in a subject by administering a composition that decreases activity or expression of a gene involved in the pathogenesis of two or more gastrointestinal viruses. More particularly, the present invention provides a method of decreasing an unspecified gastrointestinal infection in a subject comprising: a) diagnosing a subject with an unspecified gastrointestinal infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by two or more gastrointestinal viruses selected from the group consisting of a flavivirus, a filovirus, a calcivirus or a reovirus. As set forth above, in the methods of the present invention, the two or more gastrointestinal viruses can be from the same family or from a different family of gastrointestinal viruses. More particularly, and not to be limiting, the gastrointestinal virus can be any strain of reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus. In this method, the composition can be a composition that inhibits infection by three or more, four or more, five or more; or six or more gastrointestinal viruses selected from the group consisting of a flavivirus, a filovirus, a calcivirus or a reovirus.


The present invention also provides a method of preventing or decreasing an unspecified pandemic or bioterror threat in a subject comprising: a) diagnosing a subject with an unspecified pandemic or bioterrorist inflicted infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition inhibits infection by two or more, three or more, four or more; or five or more viruses selected from the group consisting of a pox virus, an influenza virus, West Nile virus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus and a Dengue fever virus.




















TABLE 3





Gene ID
Gene
Flu
RSV
HSV2
Cowpox
R-16
Dengue
HIV
HCV
WNV
Spectrum


























196
AHR



X





1


1601
DAB2
X
X
X
X
X




5


55157
DARS2
X
X
X
X
X




5


1915
EEF1A1
X
X
X
X
X

X

X
7


2023
ENO1


X
X
X
X



4


26762
HAVCR1
X

X
X
X
X



5


3190
HNRNPK
X
X
X
X
X




5


3688
ITGB1


X

X
X



3


4010
LMX1B
X

X

X
X



4


222484
LNX2
X
X
X

X

X


5


4134
MAP4

X
X



X


3


81788
NUAK2
X
X
X

X


X

5


5094
PCBP2


X


X

X

3


5429
POLH

X

X
X
X



4


5829
PXN
X
X

X

X


X
5


6122
RPL3
X

X
X
X
X



5


6222
RPS18
X
X
X
X
X
X



6


6428
SFRS3

X
X
X
X
X



5


94081
SFXN1


X
X

X



3


219938
SPATA19

X
X

X
X



4


84620
ST6GAL2

X
X

X
X



4


81671
TMEM49

X
X
X





3


84614
ZBTB37

X
X
X





3


23211
ZC3H4
X
X







2


389114
ZNF662

X
X
X
X
X



5









Combinations of gene products can be inhibited in a cell or in a subject to achieve inhibition of two or more, three or more, four or more, five or more, six or more, seven or more viruses etc. Any combination of compositions that decrease expression and/or activity of two or more, three or more, four or more, five or more, six or more gene products set forth in Table 1 can be administered to inhibit infection by two or more, three or more, four or more, five or more or six or more viruses.


Also provided by the present invention is a method of managing secondary infections in a patient comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, wherein the composition can inhibit infection by HIV and one or more, two or more, three or more, four or more; or five or more secondary infections.


As set forth above, the genes set forth in Table 1 can be involved in the pathogenesis of three or more pathogens. Therefore, the present invention provides methods of treating or preventing an unspecified infection by administering a composition that decreases the activity or expression of a gene that is involved in the pathogenesis of three or more pathogens. Therefore, the present invention provides a method of decreasing infection in a subject comprising: a) diagnosing a subject with an unspecified infection and; b) administering a composition that decreases the expression or activity of a gene or gene product set forth in Table 1, wherein the composition decreases infection by three or more pathogens. More specifically, the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


The infection can be a viral infection, a parasitic infection, a bacterial infection or a fungal infection, to name a few. As utilized herein, “an unspecified infection” is an infection that presents symptoms associated with an infection, but is not identified as specific infection. One of skill in the art, for example, a physician, a nurse, a physician's assistant, a medic or any other health practitioner would know how to diagnose the symptoms of infection even though the actual pathogen may not be known. For example, the patient can present with one or more symptoms, including, but not limited to, a fever, fatigue, lesions, weight loss, inflammation, a rash, pain (for example, muscle ache, headache, ear ache, joint pain, etc.), urinary difficulties, respiratory symptoms (for example, coughing, bronchitis, lung failure, breathing difficulties, bronchiolitis, airway obstruction, wheezing, runny nose, sinusitis, congestion, etc.), gastrointestinal symptoms (for example, nausea, diarrhea, vomiting, dehydration, abdominal pain, intestinal cramps, rectal bleeding, etc.), This can occur in the event of a bioterrorist attack or a pandemic. In this event, one of skill in the art would know to administer a composition that inhibits infection by decreasing the expression or activity of a gene or gene product set forth in Table 1 that is involved in the pathogenesis of several pathogens. Similarly, if there is a threat of an unspecified infection, for example, a threat of a bioterrorist attack, a composition that decreases the expression or activity of a gene or gene product set forth in Table 1 can be administered prophylactically to a subject to prevent an unspecified infection in a subject.


By “treat,” “treating,” or “treatment” is meant a method of reducing the effects of an existing infection. Treatment can also refer to a method of reducing the disease or condition itself rather than just the symptoms. The treatment can be any reduction from native levels and can be, but is not limited to, the complete ablation of the disease or the symptoms of the disease. Treatment can range from a positive change in a symptom or symptoms of viral infection to complete amelioration of the viral infection as detected by art-known techniques. For example, a disclosed method is considered to be a treatment if there is about a 10% reduction in one or more symptoms of the disease in a subject with the disease when compared to native levels in the same subject or control subjects. Thus, the reduction can be about a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.


The methods of the present invention can also result in a decrease in the amount of time that it normally takes to see improvement in a subject. For example, a decrease in infection can be a decrease of hours, a day, two days, three days, four days, five days, six days, seven days, eight days, nine days, ten days, eleven days, twelve days, thirteen days, fourteen days, fifteen days or any time in between that it takes to see improvement in the symptoms, viral load or any other parameter utilized to measure improvement in a subject. For example, if it normally takes 7 days to see improvement in a subject not taking the composition, and after administration of the composition, improvement is seen at 6 days, the composition is effective in decreasing infection. This example is not meant to be limiting as one of skill in the art would know that the time for improvement will vary depending on the infection.


As utilized herein, by “prevent,” “preventing,” or “prevention” is meant a method of precluding, delaying, averting, obviating, forestalling, stopping, or hindering the onset, incidence, severity, or recurrence of infection. For example, the disclosed method is considered to be a prevention if there is about a 10% reduction in onset, incidence, severity, or recurrence of infection, or symptoms of infection (e.g., inflammation, fever, lesions, weight loss, etc.) in a subject exposed to an infection when compared to control subjects exposed to an infection that did not receive a composition for decreasing infection. Thus, the reduction in onset, incidence, severity, or recurrence of infection can be about a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to control subjects. For example, and not to be limiting, if about 10% of the subjects in a population do not become infected as compared to subjects that did not receive preventive treatment, this is considered prevention.


Also provided is a method of decreasing infection in a subject comprising: a) administering a composition that decreases the expression or activity of a gene or gene product set forth in Table 1 in a subject with an unspecified infection; b) diagnosing the type of infection in the subject and; c) administering a composition that decreases the expression or activity of a gene or a gene product set forth in Table 1 for the diagnosed infection. Further provided is a method of treating viral infection comprising: a) diagnosing a subject with a viral infection; and b) removing a drug from the subject that decreases the expression or activity of a gene or gene product set forth in Table 1, if the viral infection is not a viral infection that is inhibited by a composition that decreases the expression or activity of a gene or gene product set forth in Table 1. As mentioned above, upon recognizing that a subject has an infection or the symptoms of an infection, for example, in the case of a bioterrorist attack or a pandemic, given that a gene or gene product set forth in Table 1 can be involved in the pathogenesis of several pathogens, a practitioner can prescribe or administer a composition that decreases the expression or activity of the gene or gene product. After administration, the practitioner, who can be the same practitioner or a different practitioner, can diagnose the type of infection in a subject. This diagnosis can be a differential diagnosis where the practitioner distinguishes between infections by comparing signs or symptoms and eliminates certain types of infection before arriving at the diagnosis for a specific infection, or a diagnosis based on a test that is specific for a particular infection. Once a specific infection is diagnosed, if the gene or gene product is involved in the pathogenesis of this infection, the practitioner can prescribe or administer a composition that decreases the expression or activity of that gene or gene product. This can be the same composition administered prior to diagnosis of the specific infection or a different composition that decreases expression or activity.


Also provided is a method of preventing infection in a subject comprising administering to a subject susceptible to an unspecified infection a composition that decreases the expression or activity of a gene or gene product set forth in Table 1. The composition can be administered in response to a lethal outbreak of an infection. For example, the infection can be a pandemic or a bioterrorist created infection. If there is a threat of an unspecified infection, such as a viral infection, a bacterial infection, a parasitic infection or an infection by a chimeric pathogenic agent, to name a few, a composition can be administered prophylactically to a subject to prevent an unspecified infection in a subject. The threat can also come in the form of a toxin. One of skill in the art would know to administer a composition that inhibits infection by decreasing the expression or activity of any gene or gene product set forth in Table 1 that is involved in the pathogenesis of two or more, three ore more, four or more; or five or more pathogens.


Such prophylactic use can decrease the number of people in a population that are infected, thus preventing further spread of a pandemic or decreasing the effects of a bioterrorist attack.


The composition(s) can be administered before or after infection. The decrease in infection in a subject need not be complete as this decrease can be a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or any other percentage decrease in between as long as a decrease occurs. This decrease can be correlated with amelioration of symptoms associated with infection. These compositions can be administered to a subject alone or in combination with other therapeutic agents described herein, such as anti-viral compounds, antibacterial agents, antifungal agents, antiparasitic agents, anti-inflammatory agents, anti-cancer agents, etc. Examples of viral infections, bacterial infections, fungal infections parasitic infections are set forth above. The compounds set forth herein or identified by the screening methods set forth herein can be administered to a subject to decrease infection by any pathogen or infectious agent set forth herein. Any of the compounds set forth herein or identified by the screening methods of the present invention can also be administered to a subject to decrease infection by any pathogen, now known or later discovered in which a gene in Table 1 is involved.


In the methods of the present invention, the composition can comprise one or more of, a chemical, a compound, a small molecule, an inorganic molecule, an aptamer, an organic molecule, a drug, a protein, a cDNA, a peptide, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an miRNA, an antisense nucleic acid or a ribozyme that decreases the expression or activity of a gene or gene product set forth in Table 1. The composition can be administered before or after infection. The decrease in infection in a subject need not be complete as this decrease can be a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or any other percentage decrease in between as long as a decrease occurs. This decrease can be correlated with amelioration of symptoms associated with infection. These compositions can be administered to a subject alone or in combination with other therapeutic agents described herein, such as anti-viral compounds, antibacterial agents, antifungal agents, antiparasitic agents, anti-inflammatory agents, anti-cancer agents, etc. Examples of viral infections, bacterial infections, fungal infections parasitic infections are set forth above. The compounds set forth herein or identified by the screening methods set forth herein can be administered to a subject to decrease infection by any pathogen or infectious agent set forth herein. Any of the compounds set forth herein or identified by the screening methods of the present invention can also be administered to a subject to decrease infection by any pathogen, now known or later discovered in which a gene or gene product set forth in Table 1 is involved.


Various delivery systems for administering the therapies disclosed herein are known, and include encapsulation in liposomes, microparticles, microcapsules, expression by recombinant cells, receptor-mediated endocytosis (Wu and Wu, J. Biol. Chem. 1987, 262:4429-32), and construction of therapeutic nucleic acids as part of a retroviral or other vector. Methods of introduction include, but are not limited to, mucosal, topical, intradermal, intrathecal, intratracheal, via nebulizer, via inhalation, intramuscular, otic delivery (ear), eye delivery (for example, eye drops), intraperitoneal, vaginal, rectal, intravenous, subcutaneous, intranasal, and oral routes. The compounds can be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (for example, oral mucosa, rectal, vaginal and intestinal mucosa, etc.) and can be administered together with other biologically active agents. Administration can be systemic or local. Pharmaceutical compositions can be delivered locally to the area in need of treatment, for example by topical application or local injection.


Pharmaceutical compositions are disclosed that include a therapeutically effective amount of a RNA, DNA, antisense molecule, ribozyme, siRNA, shRNA molecule, miRNA molecule, aptamer, drug, protein, small molecule, peptide inorganic molecule, organic molecule, antibody or other therapeutic agent, alone or with a pharmaceutically acceptable carrier. Furthermore, the pharmaceutical compositions or methods of treatment can be administered in combination with (such as before, during, or following) other therapeutic treatments, such as other antiviral agents, antibacterial agents, antifungal agents and antiparasitic agents.


For all of the administration methods disclosed herein, each method can optionally comprise the step of diagnosing a subject with an infection or diagnosing a subject in need of prophylaxis or prevention of infection.


Delivery Systems

The pharmaceutically acceptable carriers useful herein are conventional. Remington's Pharmaceutical Sciences, by Martin, Mack Publishing Co., Easton, Pa., 15th Edition (1975), describes compositions and formulations suitable for pharmaceutical delivery of the therapeutic agents herein disclosed. In general, the nature of the carrier will depend on the mode of administration being employed. For instance, parenteral formulations usually include injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, sesame oil, glycerol, ethanol, combinations thereof, or the like, as a vehicle. The carrier and composition can be sterile, and the formulation suits the mode of administration. In addition to biologically-neutral carriers, pharmaceutical compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.


The composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder. For solid compositions (for example powder, pill, tablet, or capsule forms), conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, sodium saccharine, cellulose, magnesium carbonate, or magnesium stearate. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.


Embodiments of the disclosure including medicaments can be prepared with conventional pharmaceutically acceptable carriers, adjuvants and counterions as would be known to those of skill in the art.


The amount of therapeutic agent effective in decreasing or inhibiting infection can depend on the nature of the pathogen and its associated disorder or condition, and can be determined by standard clinical techniques. Therefore, these amounts will vary depending on the type of virus, bacteria, fungus, parasite or other pathogen. For example, the dosage can be anywhere from 0.01 mg/kg to 100 mg/kg. Multiple dosages can also be administered depending on the type of pathogen, and the subject's condition. In addition, in vitro assays can be employed to identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.


The disclosure also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. Instructions for use of the composition can also be included.


In an example in which a nucleic acid is employed to reduce infection, such as an antisense or siRNA molecule, the nucleic acid can be delivered intracellularly (for example by expression from a nucleic acid vector or by receptor-mediated mechanisms), or by an appropriate nucleic acid expression vector which is administered so that it becomes intracellular, for example by use of a retroviral vector (see U.S. Pat. No. 4,980,286), or by direct injection, or by use of microparticle bombardment (such as a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (for example Joliot et al., Proc. Natl. Acad. Sci. USA 1991, 88:1864-8). siRNA carriers also include, polyethylene glycol (PEG), PEG-liposomes, branched carriers composed of histidine and lysine (HK polymers), chitosan-thiamine pyrophosphate carriers, surfactants (for example, Survanta and Infasurf), nanochitosan carriers, and D5W solution. The present disclosure includes all forms of nucleic acid delivery, including synthetic oligos, naked DNA, plasmid and viral delivery, integrated into the genome or not.


As mentioned above, vector delivery can be via a viral system, such as a retroviral vector system which can package a recombinant retroviral genome (see e.g., Pastan et al., Proc. Natl. Acad. Sci. U.S.A. 85:4486, 1988; Miller et al., Mol. Cell. Biol. 6:2895, 1986).


The recombinant retrovirus can then be used to infect and thereby deliver to the infected cells a nucleic acid, for example an antisense molecule or siRNA. The exact method of introducing the altered nucleic acid into mammalian cells is, of course, not limited to the use of retroviral vectors. Other techniques are widely available for this procedure including the use of adenoviral vectors (Mitani et al., Hum. Gene Ther. 5:941-948, 1994), adeno-associated viral (AAV) vectors (Goodman et al., Blood 84:1492-1500, 1994), lentiviral vectors (Naidini et al., Science 272:263-267, 1996), and pseudotyped retroviral vectors (Agrawal et al., Exper. Hematol. 24:738-747, 1996). Other nonpathogenic vector systems such as the foamy virus vector can also be utilized (Park et al. “Inhibition of simian immunodeficiency virus by foamy virus vectors expressing siRNAs.” Virology. 2005 Sep. 20). It is also possible to deliver short hairpin RNAs (shRNAs) via vector delivery systems in order to inhibit gene expression (See Pichler et al. “In vivo RNA interference-mediated ablation of MDR1 P-glycoprotein.” Clin Cancer Res. 2005 Jun. 15; 11(12):4487-94; Lee et al. “Specific inhibition of HIV-1 replication by short hairpin RNAs targeting human cyclin T1 without inducing apoptosis.” FEBS Lett. 2005 Jun. 6; 579(14):3100-6.).


Physical transduction techniques can also be used, such as liposome delivery and receptor-mediated and other endocytosis mechanisms (see, for example, Schwartzenberger et al., Blood 87:472-478, 1996) to name a few examples. This invention can be used in conjunction with any of these or other commonly used gene transfer methods.


Transgenic Cells and Non-Human Mammals

The present invention also provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 1, wherein the mammal has decreased susceptibility to infection by a pathogen, such as a virus, a bacterium, a fungus or a parasite. Exemplary transgenic non-human mammals include, but are not limited to, ferrets, fish, guinea piags, chinchilla, mice, monkeys, rabbits, rats, chickens, cows, and pigs. Such knock-out animals are useful for reducing the transmission of viruses from animals to humans and for further validating a target. In the transgenic animals of the present invention one or both alleles of a gene set forth in Table 1 can be functionally deleted.


The present invention also provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 1 wherein the mammal has decreased susceptibility to infection by two or more, three or more, four or more, or five or more pathogens selected from the group consisting of a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, an adenovirus, a flavivirus, a filovirus, a calicivirus or a reovirus. The two or more, three or more, four or more; or five or more pathogens can be respiratory viruses selected from the group consisting of Franciscella tularensis, influenza, RSV, rhinovirus, parainfluenza virus, pox virus, and measles. The two or more, three or more, four or more; or five or more pathogens can be gastrointestinal viruses selected from the group consisting of a reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a Dengue fever virus, a West Nile virus, a yellow fever virus, a rotavirus and an enterovirus. The two or more, three or more, four or more; or five or more pathogens can be selected from the group consisting of Franciscela tularensis, HIV, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, BVDV, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.


By “decreased susceptibility” is meant that the animal is less susceptible to infection or experiences decreased infection by a pathogen as compared to an animal that does not have one or both alleles of a a gene set forth in Table 1 functionally deleted. The animal does not have to be completely resistant to the pathogen. For example, the animal can be 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or any percentage in between less susceptible to infection by a pathogen as compared to an animal that does not have a functional deletion of a gene set forth in Table 1. Furthermore, decreasing infection or decreasing susceptibility to infection includes decreasing entry, replication, pathogenesis, insertion, lysis, or other steps in the replication strategy of a virus or other pathogen into a cell or subject, or combinations thereof.


Therefore, the present invention provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 1, wherein the mammal has decreased susceptibility to infection by a pathogen, such as a virus, a bacterium, a parasite or a fungus. A functional deletion is a mutation, partial or complete deletion, insertion, or other variation made to a gene sequence that inhibits production of the gene product or renders a gene product that is not completely functional or non-functional. Functional deletions can be made by insertional mutagenesis (for example via insertion of a transposon or insertional vector), by site directed mutagenesis, via chemical mutagenesis, via radiation or any other method now known or developed in the future that results in a transgenic animal with a functional deletion of a gene set forth in Table 1.


Alternatively, a nucleic acid sequence such as siRNA, a morpholino or another agent that interferes with a gene set forth in Table 1 can be delivered. The expression of the sequence used to knock-out or functionally delete the desired gene can be regulated by an appropriate promoter sequence. For example, constitutive promoters can be used to ensure that the functionally deleted gene is not expressed by the animal. In contrast, an inducible promoter can be used to control when the transgenic animal does or does not express the gene of interest. Exemplary inducible promoters include tissue-specific promoters and promoters responsive or unresponsive to a particular stimulus (such as light, oxygen, chemical concentration, such as a tetracycline inducible promoter).


The transgenic animals of the present invention that comprise a functionally deleted a gene set forth in Table 1 can be examined during exposure to various pathogens. Comparison data can provide insight into the life cycles of pathogens. Moreover, knock-out animals or functionally deleted (such as birds or pigs) that are otherwise susceptible to an infection (for example influenza) can be made to resist infection, conferred by disruption of the gene. If disruption of the gene in the transgenic animal results in an increased resistance to infection, these transgenic animals can be bred to establish flocks or herds that are less susceptible to infection.


Transgenic animals, including methods of making and using transgenic animals, are described in various patents and publications, such as WO 01/43540; WO 02/19811; U.S. Pub. Nos: 2001-0044937 and 2002-0066117; and U.S. Pat. Nos. 5,859,308; 6,281,408; and 6,376,743; and the references cited therein.


The transgenic animals of this invention also include conditional gene knockdown animals produced, for example, by utilizing the SIRIUS-Cre system that combines siRNA for specific gene-knockdown, Cre-loxP for tissue-specific expression and tetracycline-on for inducible expression. These animals can be generated by mating two parental lines that contain a specific siRNA of interest gene and tissue-specific recombinase under tetracycline control. See Chang et al. “Using siRNA Technique to Generate Transgenic Animals with Spatiotemporal and Conditional Gene Knockdown.” American Journal of Pathology 165: 1535-1541 (2004) which is hereby incorporated in its entirety by this reference regarding production of conditional gene knockdown animals.


The present invention also provides cells including an altered or disrupted gene set forth in Table 1 that are resistant to infection by a pathogen. These cells can be in vitro, ex vivo or in vivo cells and can have one or both alleles altered. These cells can also be obtained from the transgenic animals of the present invention. Such cells therefore include cells having decreased susceptibility to a virus or any of the other pathogens described herein, including bacteria, parasites and fungi.


Since the genes set forth herein are involved in viral infection, also provided herein are methods of overexpressing any of the genes set forth in Table 1 in host cells.


Overexpression of these genes can provide cells that increase the amount of virus produced by the cell, thus allowing more efficient production of viruses. Also provided is the overexpression of the genes set forth herein in avian eggs, for example, in chicken eggs.


Methods of screening agents, such as a chemical, a compound, a small or large molecule, an organic molecule, an inorganic molecule, a peptide, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an miRNA, an antisense nucleic acid or a ribozyme set forth using the transgenic animals described herein are also provided.


Screening for Resistance to Infection

Also provided herein are methods of screening host subjects for resistance to infection by characterizing a nucleotide sequence or amino acid sequence of a host gene set forth in Table 1. The nucleic acid or amino acid sequence of a subject can be isolated, sequenced, and compared to the wildtype sequence of a gene set forth in Table 1. The greater the similarity between that subject's nucleic acid sequence or amino acid sequence and the wildtype sequence, the more susceptible that person is to infection, while a decrease in similarity between that subject's nucleic acid sequence or amino acid sequence and the wildtype sequence, the more resistant that subject can be to infection. Such screens can be performed for any gene set forth in Table 1 for any species.


Assessing the genetic characteristics of a population can provide information about the susceptibility or resistance of that population to viral infection. For example, polymorphic analysis of alleles in a particular human population, such as the population of a particular city or geographic area, can indicate how susceptible that population is to infection. A higher percentage of alleles substantially similar to a wild-type gene set forth in Table 1 can indicate that the population is more susceptible to infection, while a large number of polymorphic alleles that are substantially different than a wild-type gene sequence can indicate that a population is more resistant to infection. Such information can be used, for example, in making public health decisions about vaccinating susceptible populations.


The present invention also provides a method of screening a cell for a variant form of a gene set forth in Table 1. A variant can be a gene with a functional deletion, mutation or alteration in the gene such that the amount or activity of the gene product is altered. These cells containing a variant form of a gene can be contacted with a pathogen to determine if cells comprising a naturally occurring variant of a gene set forth in Table 1 differs in their resistance to infection. For example, cells from an animal, for example, a chicken, can be screened for a variant form of a gene set forth in Table 1. If a naturally occurring variant is found and chickens possessing a variant form of the gene in their genome are less susceptible to infection, these chickens can be selectively bred to establish flocks that are resistant to infection. By utilizing these methods, flocks of chickens that are resistant to avian flu or other pathogens can be established. Similarly, other animals can be screened for a variant form of a gene set forth in Table 1. If a naturally occurring variant is found and animals possessing a variant form of the gene in their genome are less susceptible to infection, these animals can be selectively bred to establish populations that are resistant to infection. These animals include, but are not limited to, cats, dogs, livestock (for example, cattle, horses, pigs, sheep, goats, etc.), laboratory animals (for example, mouse, monkey, rabbit, rat, gerbil, guinea pig, etc.) and avian species (for example, flocks of chickens, geese. turkeys, ducks, pheasants, pigeons, doves etc.). Therefore, the present application provides populations of animals that comprise a naturally occurring variant of a gene set forth in Table 1 that results in decreased susceptibility to viral infection, thus providing populations of animals that are less susceptible to viral infection. Similarly, if a naturally occurring variant is found and animals possessing a variant form of the gene in their genome are less susceptible to bacterial, parasitic or fungal infection, these animals can be selectively bred to establish populations that are resistant to bacterial, parasitic or fungal infection.


Also provided is a method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a protein from Table 1; c) contacting the cell with an infectious pathogen; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; e) associating the agent with decreasing expression or activity of a protein from Table 1.


This method can further comprise making the association by measuring the level of expression and/or activity of a protein from Table 1.


Further provided is a method of making a compound that decreases infection in a cell by a pathogen, comprising: a) optimizing a compound to bind a protein from Table 1; b) administering the compound to a cell containing a cellular gene encoding a protein from Table 1; c) contacting the cell with an infectious pathogen; d) determining the level of infection, a decrease or elimination of infection indicating the making of a compound that decreases infection in a cell by a pathogen. This method can further comprise making a compound that decreases infection in a cell by a pathogen comprising synthesizing therapeutic quantities of the compound made.


The present invention also provides a method of synthesizing a compound that binds to a gene product of Table 1 and decreases infection by a pathogen comprising: a) contacting a library of compounds with a gene product of Table 1; b) associating binding with a decrease in infection; and c) synthesizing derivatives of the compounds from the library that bind to the gene product of Table 1.


Further provided is a business method to reduce the cost of discovery of drugs that can reduce infection by a pathogen comprising: a) screening, outside of the United States, for drugs that reduce infection by binding to or reducing the function of a gene product of Table 1; and b) importing active drugs into the United States.


Also provided is a method of making drugs comprising directing the synthesis of drugs that reduce infection by binding to or reducing the function of a gene or gene product of Table 1.


The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the antibodies, polypeptides, nucleic acids, compositions, and/or methods claimed herein are made and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for.


EXAMPLES

Following infection with the U3NeoSV1 retrovirus gene trap shuttle vector, libraries of mutagenized Vero cells were isolated in which each clone contained a single gene disrupted by provirus integration. Gene entrapment was performed essentially as described in U.S. Pat. No. 6,448,000 and U.S. Pat. No. 6,777,177. The entrapment libraries were infected with HSV, RSV, rhinovirus or Dengue fever virus and virus-resistant clones were selected as described below.


HSV

Four days prior to infection, Vero gene trap library cells were thawed at room temperature. 13 mLs of complete growth medium and a thawed gene trap library aliquot were combined in a sterile 15 mL conical tube. This was centrifuged at 1000 rpm for 5 minutes to pellet the cells. The supernatant was discarded and the cells were resuspended in complete growth medium and the aliquot of cells seeded into 4 T150 flask. The cells were allowed to grow for 4 days at 37° C. in 5% CO2 or until the cells were 70-100% confluent. On the day of infection, the medium in the T150 flasks was replaced with 19 mLs of fresh complete growth medium immediately before infecting the cells. One aliquot of HSV Strain 186 was thawed from the −80° C. freezer at 4° C. for 30 minutes. The HSV-2 (186 strain) was diluted in complete growth medium to a final concentration of 495 p.f.u./ml. 1 mL of diluted virus was added to each of the 4 T150 flasks containing Vero gene trap library cells. The cells were incubated at 37° C., 5% CO2 for 2 hours. The medium was discarded from the flasks into the waste container and replaced with 20 mLs of fresh complete growth medium to remove the inoculum. The cells were incubated at 37° C., 5% CO2. Infection was allowed to proceed without changing the medium until the cells were approximately 90% dead or dying (routinely 3 or 4 days post-infection). From then on, the medium was changed daily through day 7 post-infection. The medium was changed on days 10, 14, 17, 21, etc. post-infection. HSV-resistant colonies (clones) were observed 2-3 weeks post-infection by examining the under side of the flasks. When visible colonies appeared, they were marked and looked at under the microscope to determine which colonies are either (A) unhealthy/dying cells or are (B) actually two colonies very close together. 24-well plate(s) with 1 mL of complete growth medium in as many wells as there were resistant colonies were prepared. Resistant cells were trypsinized and cells from each HSV-resistant clone were transferred to a single well of the 24 well plate (already containing 1 ml of complete growth medium). This process was repeated for each colony. The colonies were grown until cells in several wells approach 20-30% confluency. At this point, cells were detached and seeded into duplicate 24-well plates. Resistance confirmation was performed by re-infecting clones in one 24-well plate. Following identification of resistant clones, resistant clones in the uninfected 24-well plates were expanded in T75 flasks for subsequent genomic DNA isolation (DNeasy kits, Qiagen, Inc.).


Identification of Genes Disrupted in HSV-Resistant Clones

The U3NeoSV1 gene trap vector contains a plasmid origin of replication and ampicillin resistance gene; thus, regions of genomic DNA adjacent to the targeting vector were readily cloned by plasmid rescue and sequenced. The flanking sequences were compared to the nucleic acid databases to identify candidate cellular genes that confer resistance to lytic infection by herpes simplex virus when altered by gene entrapment.


RSV

Four days prior to infection, Vero gene trap library cells were thawed at room temperature. 13 mLs of complete growth medium and a thawed gene trap library aliquot were combined in a sterile 15 mL conical tube. This was centrifuged at 1000 rpm for 5 minutes to pellet the cells. The supernatant was discarded and the cells were resuspended in complete growth medium and the aliquot of cells seeded into 4 T150 flask. The cells were allowed to grow for 4 days at 37° C. in 5% CO2 or until the cells were 70-100% confluent. On the day of infection, the medium in the T150 flasks was replaced with 19 mLs of fresh complete growth medium immediately before infecting the cells. One aliquot of RSV A2 strain was thawed from the −80° C. freezer at 4° C. for 30 minutes. The


RSV A2 strain was diluted in complete growth medium to a final concentration of 11,812 p.f.u./ml. 1 mL of diluted virus was added to each of the 4 T150 flasks containing Vero gene trap library cells. The cells were incubated at 37° C., 5% CO2 for 2 hours. The medium was discarded from the flasks and replaced with 20 mLs of fresh complete growth medium to remove the inoculum. The cells were incubated at 37° C., 5% CO2. Infection was allowed to proceed without changing the medium until the cells were approximately 90% dead or dying (approximately 3 or 4 days post-infection). From then on, the medium was changed daily through day 7 post-infection. The medium was changed on days 10, 14, 17, 21, etc. post-infection. RSV-resistant colonies (clones) were observed 2-3 weeks post-infection by examining the under side of the flasks. When visible colonies appeared, they were marked and looked at under the microscope to determine which colonies are either (A) unhealthy/dying cells or are (B) actually two colonies very close together. 24-well plate(s) with 1 mL of complete growth medium in as many wells as there were resistant colonies were prepared. Resistant cells were trypsinized and cells from each RSV-resistant clone were transferred to a single well of the 24 well plate (already containing 1 ml of complete growth medium). This process was repeated for each colony. The colonies were grown until cells in several wells approach 20-30% confluency. At this point, cells were detached and seeded into duplicate 24-well plates. Resistance confirmation was performed by re-infecting clones in one 24-well plate. Following identification of resistant clones, resistant clones in the uninfected 24-well plates were expanded in T75 flasks for subsequent genomic DNA isolation (DNeasy kits, Qiagen, Inc.).


Identification of Genes Disrupted in RSV-Resistant Clones

The U3NeoSV1 gene trap vector contains a plasmid origin of replication and ampicillin resistance gene; thus, regions of genomic DNA adjacent to the targeting vector were readily cloned by plasmid rescue and sequenced. The flanking sequences were compared to the nucleic acid databases to identify candidate cellular genes that confer resistance to lytic infection by respiratory syncytial virus when altered by gene entrapment.


Rhinovirus

Four days prior to infection, an aliquot of TZM-bl gene trap library cells were thawed at room temperature. 13 mLs of complete growth medium and thawed gene trap library aliquot were combined in a sterile 15 mL conical tube. This was centrifuged at 1000 rpm for 5 minutes to pellet the cells. The supernatant was discarded The cells were resuspended in complete growth medium and the aliquot of cells was seeded into 4 T150 flaks with reclosable lids. Cells were allowed grow for 4 days at 37° C. in 5% CO2 or until the cells were 70-100% confluent. On the day of infection, the medium was replaced in the T150 flasks with 19 mLs of fresh complete growth medium immediately before infecting the cells. One aliquot of Rhinovirus-16 Strain 11757 from the −80° C. freezer was thawed at 4° C. for 30 minutes Rhinovirus was diluted in complete growth medium to a final concentration of 1×105 p.f.u./ml. Approximately 1 mL of the diluted virus was added to each of 4 T150 flasks containing TZM-bl gene trap library cells. 200 uL of sterile MgC12 was added to each T150 flask (final MgC12 concentration=40 mM). The T150 flasks were placed on a rocker and incubated at 33° C., 5% CO2, rocking cells gently at the lowest setting. Infection was allowed to proceed without changing the medium until the cells were >99.9% dead or dying (routinely 6-7 days post-infection). The medium was changed and the flasks transferred to a 37° C., 5% CO2 incubator. The medium was changed on days 10, 14, 17, 21, etc. post-infection (following this pattern of days), while maintaining cells at 37° C., 5% CO2.


Rhinovirus resistant does were observed 2-3 weeks post-infection by examining the under side of the flasks. When visible colonies appeared, they were marked and looked at under the microscope to determine which colonies are either (A) unhealthy/dying cells or are (B) actually two colonies very close together. 24-well plate(s) with 1 mL of complete growth medium in as many wells as there were resistant colonies were prepared. Resistant cells were trypsinized and cells from each rhinovirus-resistant clone were transferred to a single well of the 24 well plate (already containing 1 ml of complete growth medium). This process was repeated for each colony. The colonies were grown until cells in several wells approach 20-30% confluency. At this point, cells were detached and seeded into duplicate 24-well plates. Resistance confirmation was performed by re-infecting clones in one 24-well plate. Following identification of resistant clones, resistant clones in the uninfected 24-well plates were expanded in T75 flasks for subsequent genomic DNA isolation (DNeasy kits, Qiagen, Inc.).


Identification of Genes Disrupted in Rhinovirus-Resistant Clones

The U3NeoSV1 gene trap vector contains a plasmid origin of replication and ampicillin resistance gene; thus, regions of genomic DNA adjacent to the targeting vector were readily cloned by plasmid rescue and sequenced. The flanking sequences were compared to the nucleic acid databases to identify candidate cellular genes that confer resistance to lytic infection by rhinovirus when altered by gene entrapment.


siRNA and Small Molecule Studies


Any of the genes set forth in Table 1 is further analyzed by contacting cells comprising a gene set forth in Table 1 with siRNA or small molecule that targets the gene product of the gene, and any pathogen set forth herein to identify the gene as a gene involved in pathogenic infection (for example, and not to be limiting, an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, West Nile virus, Chikungunya virus or a Dengue fever virus). A decrease in viral infection indicates that the gene is a gene that is involved in pathogenic infection. This process can be performed for all of the genes set forth herein with any of the viruses, bacteria, parasites or fungi set forth herein.


siRNA Transfections can be performed as follows: Pools of 4 duplexed siRNA molecules targeting a gene of interest are reconstituted to a final working concentration of 50 uM as directed by the manufacturer (Qiagen). Twenty-four hours prior to transfection, cells are plated in 6-well dishes at 3×105 cells per well, such that at the time of transfection, the cells are approximately 30% confluent. Prior to transfection, the cells are washed once with 1× phosphate buffered saline, and the medium replaced with approximately 1.8 ml antibiotic-free medium. siRNA aliquots are diluted with Opti-MEM and RNAseOUT (Invitrogen), 100u1 and 1 ul per transfection, respectively. In a separate tube, transfection reagent Lipofectamine-2000 (Invitrogen) or Oligofectamine (Invitrogen) are diluted in Opti-MEM as directed by the manufacturer. Following a 5 minute incubation at room temperature, the diluted siRNA is added to the transfection reagent mixture, and incubated for an additional 20 minutes prior to adding to independent wells of the 6-well dishes. Transfections are incubated at 37° C. for 48 hours without changing the medium.


Virus Infections: Following 48-hour transfection, medium is aspirated from E-well plates. Viruses are diluted in the appropriate medium and 500u1 of either virus-free medium or virus dilution is added to each well, and adsorption is allowed to occur at the appropriate temperature for 1 hour. Following adsorption, inoculum is aspirated off the cells, cells are washed once with 1× phosphate buffered saline, and 2 ml growth medium is added to the cells. The infected cells are incubated for 72 hours at the appropriate temperature prior to harvesting samples for viral titration.


Viral Genomic Extractions: Seventy-hours after inoculating cells, medium is harvested from 6-well dishes and centrifuged for 2 minutes at 10,000 rpm to remove any cellular debris. 200 ul of clarified medium is added to 25 ul Proteinase K, to which 200 ul PureLink96 Viral RNA/DNA lysis buffer (Invitrogen) is added according to the manufacturer. Samples were processed and viral genomic RNA or DNA is extracted using an epMotion 5075 robotics station (Eppendorf) and the PureLink96 Viral RNA/DNA kit (Invitrogen).


cDNA and Quantitative Real-Time PCR Reactions: 3 ul of extracted viral RNA is converted to cDNA using M-MLV reverse transcriptase (Invitrogen) and AmpliTaq Gold PCR buffer (Applied Biosystems). MgCl2, dNTPs and RNAseOUT (Invitrogen) are added to achieve a final concentration of 5 mM, 1 mM and 2 U/ul, respectively. Random hexamers (Applied Biosystems) are added to obtain 2.5 mM final concentration. Reactions are incubated at 42° C. for 1 hour, followed by heat inactivation of the reverse transcriptase at 92° C. for 10 minutes. Quantitative real-time PCR reactions are set up in 10 ul volumes using 1 ul of template cDNA or extracted viral DNA using virus-specific TaqMan probes (Applied Biosystems) and RealMasterMix (Eppendorf). 2-step reactions are allowed to proceed through 40 to 50 cycles on an ep RealPlex thermocycler (Eppendorf). Quantitative standards for real-time PCR are constructed by cloning purified amplicons into pCR2—TOPO (Invitrogen) and sequenced as necessary.


The amount of viral replication in the cells contacted with siRNA to the gene of interest is calculated and compared to the amount of viral replication in control cells that did not receive siRNA targeting the gene of interest.


Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully derive the state of the art to which this invention pertains.

Claims
  • 1. A method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of ITGB1, MALAT1, SF3B4, C11ORF54, AMOTL2, or GDF15.
  • 2. The method of claim 1, wherein infection is decreased by decreasing the replication of the pathogen.
  • 3. The method of claim 1, wherein the pathogen is a virus.
  • 4-9. (canceled)
  • 10. The method of claim 1, wherein expression or activity of ITGB1, MALAT1, SF3B4, C11ORF54, AMOTL2, or GDF15 is decreased by contacting the cell with a composition comprising a chemical, a compound, an aptamer, a small molecule, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an antisense nucleic acid, an LNA or a ribozyme.
  • 11. The method of claim 10, wherein decreasing expression of ITGB1, MALAT1, SF3B4, C11ORF54, AMOTL2, or GDF15 comprises decreasing translation of an mRNA encoding ITGB1, MALAT1, SF3B4, C11ORF54, AMOTL2, or GDF15.
  • 12. The method of claim 11, wherein the composition comprises an antisense nucleic acid that specifically hybridizes and decreases expression or activity of ITGB1, MALAT1, SF3B4, C11ORF54, AMOTL2, or GDF15.
  • 13. The method of claim 11, wherein the composition comprises an siRNA that decreases expression or activity of ITGB1, MALAT1, SF3B4, C11ORF54, AMOTL2, or GDF15.
  • 14. The method of claim 10, wherein the composition comprises an antibody that specifically binds to a protein encoded by ITGB1, MALAT1, SF3B4, C11ORF54, AMOTL2, or GDF15.
  • 15-46. (canceled)
  • 47. A cell comprising an altered or disrupted nucleic acid encoding ITGB1, MALAT1, SF3B4, C11ORF54, AMOTL2, or GDF15, wherein the cell has decreased susceptibility to infection by a pathogen.
  • 48. The cell of claim 47, wherein the pathogen is a virus and the cell is infected with a virus.
  • 49. The cell of claim 48, wherein the virus is a respiratory virus.
  • 50. The cell of claim 49, wherein the respiratory virus is a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, or an adenovirus.
  • 51. The cell of claim 50, wherein the respiratory virus is selected from the group consisting of influenza virus, a pox virus, parainfluenza virus, adenovirus, measles, rhinovirus, and RSV.
Parent Case Info

This application claims the benefit of U.S. Application No. 61/270,930, filed on Jul. 15, 2009, which is hereby incorporated in its entirety by this reference.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US10/42140 7/15/2010 WO 00 7/19/2013
Provisional Applications (1)
Number Date Country
61270930 Jul 2009 US