METHODS AND NUCLEIC ACIDS FOR THE ANALYSIS OF GENE EXPRESSION ASSOCIATED WITH TISSUE CLASSIFICATION

Information

  • Patent Application
  • 20090191548
  • Publication Number
    20090191548
  • Date Filed
    September 28, 2006
    18 years ago
  • Date Published
    July 30, 2009
    15 years ago
Abstract
The present application provides methods and nucleic acids the classification of a biological sample. This is achieved by the analysis of the expression status of at least one of the genes selected from Table 1 as disclosed.
Description
FIELD OF THE INVENTION

The present invention relates to human DNA sequences that exhibit tissue specific expression patterns. Particular embodiments of the invention provide methods for classifying a biological sample.


BACKGROUND

Bisulfite modification of DNA is an art-recognized tool used to assess CpG methylation status. 5-methylcytosine is the most frequent covalent base modification in the DNA of eukaryotic cells. It plays a role, for example, in the regulation of the transcription, in genetic imprinting, and in tumorigenesis. Therefore, the identification of 5-methylcytosine as a component of genetic information is of considerable interest. However, 5-methylcytosine positions cannot be identified by sequencing, because 5-methylcytosine has the same base pairing behavior as cytosine. Moreover, the epigenetic information carried by 5-methylcytosine is completely lost during, e.g., PCR amplification.


The most frequently used method for analyzing DNA for the presence of 5-methylcytosine is based upon the specific reaction of bisulfite with cytosine whereby, upon subsequent alkaline hydrolysis, cytosine is converted to uracil, which corresponds to thymine in its base pairing behavior. Significantly, however, 5-methylcytosine remains unmodified under these conditions. Consequently, the original DNA is converted in such a manner that methylcytosine, which originally could not be distinguished from cytosine by its hybridization behavior, can now be detected as the only remaining cytosine using standard, art-recognized molecular biological techniques, for example, by amplification and hybridization, or by sequencing. All of these techniques are based on differential base pairing properties, which can now be fully exploited.


The prior art, in terms of sensitivity, is defined by a method comprising enclosing the DNA to be analyzed in an agarose matrix, thereby preventing the diffusion and renaturation of the DNA (bisulfite only reacts with single-stranded DNA), and replacing all precipitation and purification steps with fast dialysis (Olek A, et al., A modified and improved method for bisulfite based cytosine methylation analysis, Nucleic Acids Res. 24:5064-6, 1996). It is thus possible to analyze individual cells for methylation status, illustrating the utility and sensitivity of the method. An overview of art-recognized methods for detecting 5-methylcytosine is provided by Rein, T., et al., Nucleic Acids Res., 26:2255, 1998.


The bisulfite technique, barring few exceptions (e.g., Zeschnigk M, et al., Eur J Hum Genet. 5:94-98, 1997), is currently only used in research. In all instances, short, specific fragments of a known gene are amplified subsequent to a bisulfite treatment, and either completely sequenced (Olek and Walter, Nat Genet. 1997 17:275-6, 1997), subjected to one or more primer extension reactions (Gonzalgo and Jones, Nucleic Acids Res., 25:2529-31, 1997; WO 95/00669; U.S. Pat. No. 6,251,594) to analyze individual cytosine positions, or treated by enzymatic digestion (Xiong and Laird, Nucleic Acids Res., 25:2532-4, 1997). Detection by hybridization has also been described in the art (Olek et al., WO 99/28498). Additionally, use of the bisulfite technique for methylation detection with respect to individual genes has been described (Grigg and Clark, Bioessays, 16:431-6, 1994; Zeschnigk M, et al., Hum Mol Genet., 6:387-95, 1997; Feil R, et al., Nucleic Acids Res., 22:695-, 1994; Martin V, et al., Gene, 157:261-4, 1995; WO 97/46705 and WO 95/15373).


Methylation Assay Procedures. Various methylation assay procedures are known in the art, and can be used in conjunction with the present invention. These assays allow for determination of the methylation state of one or a plurality of CpG dinucleotides (e.g., CpG islands) within a DNA sequence. Such assays involve, among other techniques, DNA sequencing of bisulfite-treated DNA, PCR (for sequence-specific amplification), Southern blot analysis, and use of methylation-sensitive restriction enzymes.


For example, genomic sequencing has been simplified for analysis of DNA methylation patterns and 5-methylcytosine distribution by using bisulfite treatment (Frommer et al., Proc. Natl. Acad. Sci. USA 89:1827-1831, 1992). Additionally, restriction enzyme digestion of PCR products amplified from bisulfite-converted DNA is used, e.g., the method described by Sadri and Hornsby (Nucl. Acids Res. 24:5058-5059, 1996), or COBRA (Combined Bisulfite Restriction Analysis) (Xiong and Laird, Nucleic Acids Res. 25:2532-2534, 1997).


Preferably, assays such as “MethyLight™” (a fluorescence-based real-time PCR technique) (Eads et al., Cancer Res. 59:2302-2306, 1999), Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) reactions (Gonzalgo and Jones, Nucleic Acids Res. 25:2529-2531, 1997), methylation-specific PCR (“MSP”; Herman et al., Proc. Natl. Acad. Sci. USA 93:9821-9826, 1996; U.S. Pat. No. 5,786,146), are used alone or in combination with other of these methods.


MethyLight™. The MethyLight™ assay is a high-throughput quantitative methylation assay that utilizes fluorescence-based real-time PCR (TaqMan™) technology that requires no further manipulations after the PCR step (Eads et al., Cancer Res. 59:2302-2306, 1999). Briefly, the MethyLight™ process begins with a mixed sample of genomic DNA that is converted, in a sodium bisulfite reaction, to a mixed pool of methylation-dependent sequence differences according to standard procedures (the bisulfite process converts unmethylated cytosine residues to uracil). Fluorescence-based PCR is then performed either in an “unbiased” (with primers that do not overlap known CpG methylation sites) PCR reaction, or in a “biased” (with PCR primers that overlap known CpG dinucleotides) reaction. Sequence discrimination can occur either at the level of the amplification process or at the level of the fluorescence detection process, or both.


The MethyLight™ assay may be used as a quantitative test for methylation patterns in the genomic DNA sample, wherein sequence discrimination occurs at the level of probe hybridization. In this quantitative version, the PCR reaction provides for unbiased amplification in the presence of a fluorescent probe that overlaps a particular putative methylation site. An unbiased control for the amount of input DNA is provided by a reaction in which neither the primers, nor the probe overlie any CpG dinucleotides. Alternatively, a qualitative test for genomic methylation is achieved by probing of the biased PCR pool with either control oligonucleotides that do not “cover” known methylation sites (a fluorescence-based version of the “MSP” technique), or with oligonucleotides covering potential methylation sites.


The MethyLight™ process can by used with a “TaqMan®” probe in the amplification process. For example, double-stranded genomic DNA is treated with sodium bisulfite and subjected to one of two sets of PCR reactions using TaqMan® probes; e.g., with either biased primers and TaqMan® probe, or unbiased primers and TaqMan® probe. The TaqMan® probe is dual-labeled with fluorescent “reporter” and “quencher” molecules, and is designed to be specific for a relatively high GC content region so that it melts out at about 10° C. higher temperature in the PCR cycle than the forward or reverse primers. This allows the TaqMan® probe to remain fully hybridized during the PCR annealing/extension step. As the Taq polymerase enzymatically synthesizes a new strand during PCR, it will eventually reach the annealed TaqMan® probe. The Taq polymerase 5′ to 3′ endonuclease activity will then displace the TaqMan® probe by digesting it to release the fluorescent reporter molecule for quantitative detection of its now unquenched signal using a real-time fluorescent detection system.


Typical reagents (e.g., as might be found in a typical MethyLight™-based kit) for MethyLight™ analysis may include, but are not limited to: PCR primers for specific gene (or bisulfite treated DNA sequence or CpG island); TaqMan® probes; optimized PCR buffers and deoxynucleotides; and Taq polymerase.


Ms-SNuPE. The Ms-SNuPE technique is a quantitative method for assessing methylation differences at specific CpG sites based on bisulfite treatment of DNA, followed by single-nucleotide primer extension (Gonzalgo and Jones, Nucleic Acids Res. 25:2529-2531, 1997). Briefly, genomic DNA is reacted with sodium bisulfite to convert unmethylated cytosine to uracil while leaving 5-methylcytosine unchanged. Amplification of the desired target sequence is then performed using PCR primers specific for bisulfite-converted DNA, and the resulting product is isolated and used as a template for methylation analysis at the CpG site(s) of interest. Small amounts of DNA can be analyzed (e.g., microdissected pathology sections), and it avoids utilization of restriction enzymes for determining the methylation status at CpG sites.


Typical reagents (e.g., as might be found in a typical Ms-SNuPE-based kit) for Ms-SNuPE analysis may include, but are not limited to: PCR primers for specific gene (or bisulfite treated DNA sequence or CpG island); optimized PCR buffers and deoxynucleotides; gel extraction kit; positive control primers; Ms-SNuPE primers for specific gene; reaction buffer (for the Ms-SNuPE reaction); and labeled nucleotides. Additionally, bisulfite conversion reagents may include: DNA denaturation buffer; sulfonation buffer; DNA recovery reagents or kit (e.g., precipitation, ultrafiltration, affinity column); desulfonation buffer; and DNA recovery components.


MSP. MSP (methylation-specific PCR) allows for assessing the methylation status of virtually any group of CpG sites within a CpG island, independent of the use of methylation-sensitive restriction enzymes (Herman et al. Proc. Natl. Acad. Sci. USA 93:9821-9826, 1996; U.S. Pat. No. 5,786,146). Briefly, DNA is modified by sodium bisulfite converting all unmethylated, but not methylated cytosines to uracil, and subsequently amplified with primers specific for methylated versus unmethylated DNA. MSP requires only small quantities of DNA, is sensitive to 0.1% methylated alleles of a given CpG island locus, and can be performed on DNA extracted from paraffin-embedded samples. Typical reagents (e.g., as might be found in a typical MSP-based kit) for MSP analysis may include, but are not limited to: methylated and unmethylated DNA specific PCR primers for specific gene (or bisulfite treated DNA sequence or CpG island), optimized PCR buffers and deoxynucleotides, and specific probes.


Prior art of several markers. If not stated otherwise, the following information was received from the Entrez database (http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene) and the OMIM database (http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIM).


Glycoprotein Ib (platelet) beta polypeptide. (GP1BB) Glycoprotein Ib (platelet) beta polypeptide (GP1BB) is subunit of the platelet glycoprotein Ib (GPIb). GPIb is a heterodimeric transmembrane protein consisting of a disulfide-linked 140 kD alpha chain and 22 kD beta chain. It is part of the GPIb-V-IX system that constitutes the receptor for von Willebrand factor (VWF), and mediates platelet adhesion in the arterial circulation. GPIb alpha chain provides the VWF binding site, and GPIb beta contributes to surface expression of the receptor and participates in transmembrane signaling through phosphorylation of its intracellular domain. Mutations in the GPIb beta subunit have been associated with Bernard-Soulier syndrome, velocardiofacial syndrome and giant platelet disorder. The 206 amino acid precursor of GPIb beta is synthesized from a 1.0 kb mRNA expressed in platelets and megakaryocytes. A 411 amino acid protein arising from a longer, unspliced transcript in endothelial cells has been described; however, the authenticity of this product has been questioned. Yet another less abundant GPIb beta mRNA species of 3.5 kb, expressed in nonhematopoietic tissues such as endothelium, brain and heart, was shown to result from inefficient usage of a non-consensus polyA_signal within a separate gene (PNUTL1) located upstream of this gene. In the absence of polyadenylation from its own imperfect site, the PNUTL1 gene uses the consensus polyA_signal of this gene (Entrez database).


Transcription factor AP-2 alpha. Transcription factor AP-2 alpha (TFAP2A, alias AP-2 AP2TF; TFAP2; AP-2alpha) also known as activating enhancer binding protein 2 alpha is a 52-kD retinoic acid-inducible and developmentally regulated activator of transcription that binds to a consensus DNA-binding sequence CCCCAGGC in the SV40 and metallothionein (MIM 156350) promoters (OMIM database). The loss of transcription factor AP-2alpha expression has been shown to associate with tumorigenicity of melanoma cell lines and poor prognosis in primary cutaneous melanoma. Altogether these findings suggest that the gene encoding AP-2alpha (TFAP2A) acts as a tumor suppressor in melanoma. A failure in post-transcriptional processing of AP-2alpha is a possible inactivation mechanism of AP-2alpha in cutaneous melanoma (Karjalainen et al, 2000). It was shown that the induction of AP-2 mRNA is at the level of transcription and is transient, reaching a peak 48-72 hr after the addition of retinoic acid (RA) and declining thereafter, even in the continuous presence of retinoic acid. AP-2-binding site-mediated cAMP and TPA (12-O-tetradecanoyl-phorbol-13-acetate) responses are not regulated at the level of AP-2 expression but, rather, achieved either by post-translational changes in AP-2 or in conjunction with another protein (Luscher et al, 1989). Decreases in AP2 protein were rapidly reversed by insulin administration. There were no changes in AP2 protein in the absence of changes in AP2 mRNA supporting a pretranslational mechanism of regulation (Melki and Abumrad, 1993). There is a significant up-regulation of AP-2gamma expression in breast cancer specimens (P=0.01). There was also a significant correlation between the presence of the AP-2alpha protein and estrogen receptor expression (P=0.018) and between specimens containing both AP-2alpha/AP-2gamma proteins and ERBB-2 expression (P=0.003). Furthermore, we detected an association (P=0.04) between the expression of AP-2gamma and the presence of an additional signal transduction molecule implicated in breast cancer (Turner et al, 1998). Macrophages deficient in AP2 display alterations in inflammatory cytokine production (Linton and Fazio, 2003). Loss of AP-2 results in up-regulation of MCAM/MUC18 and an increase in tumor growth and metastasis of human melanoma cells (Jean et al, 1998). AP-2alpha was reduced in advanced Dukes's stage adenocarcinomas. Together with reduced AP-2gamma expression in high grade colorectal carcinomas, this might contribute to tumor progression (Ropponen et al, 2001). The developmentally regulated transcription factor AP-2 is expressed at higher levels in human fetal skeletal muscle and rhabdomyosarcoma cells compared to human adult skeletal muscle (Zhang et al, 1998). Through its distinct actions in adipocytes and macrophages, AP2 links features of the metabolic syndrome including insulin resistance, obesity, inflammation, and atherosclerosis (Linton and Fazio, 2003). Chromatin immunoprecipitation analysis demonstrated DNA binding activity of AP-2 in the TbetaRI promoter and of Sp1 in the TbetaRII promoter after treatment with 5-aza-2′-deoxycytidine (Zhang et al, 2005). Site-specific methylation in NF1 gene, involving transcription factor binding sites for SP1, CRE (−10), and AP-2, was observed. One region of the 5′-UTR (untranslated region) overlapping with a putative AP-2 binding site was methylated at 30-100% in 4/20 control samples (Harder et al, 2004). High resolution mapping of methylated cytosines revealed that differential expression of the AP-2 alpha gene in normal human lung fibroblasts and their SV40-transformed counterparts was associated with distinct patterns of cytosine methylation in the AP-2 alpha promoter just 5′ to the transcription initiation site. Site-specific methylation was positively correlated with increased AP-2 alpha gene expression in both transformed cell lines investigated (Zhu et al, 2001). High resolution mapping of methylated cytosines revealed that differential expression of the AP-2 alpha gene in normal human lung fibroblasts and their SV40-transformed counterparts was associated with distinct patterns of cytosine methylation in the AP-2 alpha promoter just 5′ to the transcription initiation site. Site-specific methylation was positively correlated with increased AP-2 alpha gene expression in both transformed cell lines investigated (Zhu et al, 2001).


Cdc42 effector protein 1. Cdc42 effector protein 1 (CDC42EP1 alias MSE55, CEP1, Borg5, MGC15316) is a member of the Rho GTPase family that regulates multiple cellular activities, including actin polymerization. The protein encoded by this gene is a CDC42 binding protein that mediates actin cytoskeleton reorganization at the plasma membrane. The encoded protein, which is secreted, is primarily found in bone marrow. Two transcript variants encoding different isoforms have been found for this gene (Entrez database). Northern blot analysis demonstrates expression limited to endothelial and bone marrow stromal cells, but not poly(A) RNA from monkey liver, spleen, brain, lung, and kidney. On this basis, this protein was designated MSE55, for marrow/stromal/endothelial cell protein with a molecular mass of 55,000 daltons. Its tissue-specific expression may suggest a functional role in hematopoiesis (Bahou et al, 1992). MSE55 induced the formation of long, actin-based protrusions in NIH 3T3 cells as detected by immunofluorescence and live-cell video microscopy. MSE55-induced protrusion formation was blocked by expression of dominant-negative N17Cdc42, but not by expression of dominant-negative N17Rac. These findings indicate that MSE55 is a Cdc42 effector protein that mediates actin cytoskeleton reorganization at the plasma membrane (Burbelo et al, 1999).


Glutathione peroxidase 5. Glutathione peroxidase 5 (GPX5) also known as glutathione reductase is part of the hydrogen peroxide scavenging system found within the epididymis in the mammalian male reproductive tract. GPX5 expression is epididymis-specific and the transcript is unique from other GPXs because it contains a deletion resulting in an mRNA that does not contain a selenocysteine (UGA) codon (an unusual amino acid present in other GPXs). This deletion also renders the mRNA incapable of encoding an active GPX isoenzyme. For this reason, GPX5 is selenium-independent and has very little activity towards hydrogen peroxide or organic hydroperoxides. GPX5, which is bound to the acrosome of sperm, may act to protect sperm from premature acrosome reaction in the epididymis (Entrez database). The cDNA of human GPX5 is cloned. Thereby it was shown that the majority of transcripts contain a 118 nt frame-shifting deletion, arising, most likely, from inappropriate excision of exon 3 during processing. Antisera raised against recombinant human GPX5 cross-reacted with rat and macaque (Macaca fascicularis) epididymal proteins of the size expected for full-length, active GPX5. However, no similar reactivity could be demonstrated in any of the human samples tested (Hall et al, 1998). The tissue-restricted polyoma enhancer activator protein (PEA3) of the ETS oncogene family of DNA-binding proteins is a putative modulator of the epididymis-specific glutathione peroxidase 5 gene GPX5 (Drevet et al, 1998). At least part of the androgenic control of the GPX5 expression is exerted at the transcriptional level via an androgen response element localized in the distal promoter region of the GPX5 gene (Lareyre et al, 1997).


Gamma-parvin. Gamma-parvin (PARVG) is a member of the parvin family, a family of actin-binding proteins associated with focal contacts (OMIM database).


NKG2D ligand 4 precursor. NKG2D ligand 4 precursor (RAET1E alias NKG2D ligand 4, NKG2DL4, N2DL-4, RL-4, LETAL, bA350J20.7, ULBP4; MGC125308; MGC125309; bA350J20.7) also known as Retinoic acid early transcript 1E, Lymphocyte effector toxicity activation ligand, RAE-1-like transcript 4 is a member of the RAET1 family. The members of this family are major histocompatibility complex (MHC) class I-related genes located within a 180-kb cluster on chromosome 6q24.2-q25.3. RAET1 proteins contain MHC class I-like alpha-1 and alpha-2 domains. RAET1E and RAET1G (MIM 609244) differ from the other RAET1 proteins (e.g., RAET1I, or ULBP1; MIM 605697) in that they have type I membrane-spanning sequences at their C termini rather than glycosylphosphatidylinositol anchor sequences (Radosavljevic et al., 2002). The expression of diverse NKG2D-binding molecules (RAET1E and RAET1G) in different tissues and with different properties is consistent with multiple modes of infection- or stress-induced activation (Bacon et al, 2004). Tissue expression of ULBP4 differs from other members of the family, in that it is expressed predominantly in the skin (Jan Chalupny et al, 2003).


Oncostatin M precursor. Oncostatin M precursor (OSM alias MGC20461) is a member of a cytokine family that includes leukemia-inhibitory factor, granulocyte colony-stimulating factor, and interleukin 6. This gene encodes a growth regulator which inhibits the proliferation of a number of tumor cell lines. It regulates cytokine production, including IL-6, G-CSF and GM-CSF from endothelial cells. The related members of the interleukin 6 (IL-6) family of cytokines, IL-6, leukemia inhibitory factor (LIF), and oncostatin M, act as major inflammatory mediators and induce the hepatic acute phase reaction. Normal parenchymal liver cells express the receptors for these cytokines, and these receptors activate, to a comparable level, the intracellular signaling through signal transducer and activator of transcription (STAT) proteins and extracellular-regulated kinase (ERK) (Entrez database). OSM stimulates the expression of the immediate early genes c-fos and Egr-1 in Gnv-4 cells, an effect dependent upon the activation of the MAPK Erk1/2 intracellular signaling pathway (Igaz et al, 2005). OSM and macrophage-derived cytokines suppressed proliferation of normal epithelial cells, but reduced inhibition or even stimulated proliferation was noted for preneoplastic cells (Loewen et al, 2005). In human liver, OSM protein is expressed in Kupffer cells, variably in normals but universally in cirrhosis. The differential expression pattern of OSM and its receptors could allow for differential OSM signaling by alternative utilization of receptors to promote hepatocyte proliferation in acute injury and, with its homologue LIF, for the bile ductular reaction in cirrhosis (Znoyko et al, 2005). OSM strongly and specifically affects the expression of many genes, in particular those involved with innate immunity, angiogenesis, adhesion, motility, tissue remodeling, cell cycle and transcription (Finelt et al, 2005). OSM has a strong lipid-lowering effect under in vivo conditions in which the levels of circulating LDL-C are high and liver LDLR transcription is repressed (King et al, 2005). Oncostatin M (OSM), a member of the interleukin-6 family of cytokines, is thought to be expressed mostly by activated T-lymphocytes and monocytes in adult animals. However, specific constitutive tissue expression is reported of OSM in the pancreas, kidney, testes, spleen, stomach, and brain, but not liver or lung, of three adult rodent species (Znoyko et al, 2005). These studies identify S100A9 as a novel OSM-regulated gene through the STAT3-signaling cascade and suggest its involvement in the growth regulation of breast cancer cells (Li et al, 2004). Oncostatin M stimulates the detachment of a reservoir of invasive mammary carcinoma cells (Holzer et al, 2004). Blood neutrophils can be stimulated to express and rapidly release large quantities of OSM. It is proposed that OSM is released from neutrophils as they infiltrate rheumatoid joints and, thus, contribute to the complex cytokine network that characterizes retinoic acid (Cross et al, 2004). Oncostatin-M may contribute to the process of healing after myocardial infarction (Gwechenberger et al, 2004). Oncostatin M expression is upregulated in cirrhosis where it may have a role as a profibrogenic cytokine in hepatic stellate cells (Levy et al, 2000). OSM induces an angiogenic effect on capillary endothelial cells, which could be, at least in part, implicated in pathological processes such as atherosclerosis or tumor growth (Vasse et al, 1999).


Cytohesin-4. Cytohesin-4 (PSCD4 alias CYT4) also known as Pleckstrin homology, Sec7 and coiled/coil domains 4 (PSCD4) is a member of the PSCD family. Members of this family have identical structural organization that consists of an N-terminal coiled-coil motif, a central Sec7 domain, and a C-terminal pleckstrin homology (PH) domain. The coiled-coil motif is involved in homodimerization, the Sec7 domain contains guanine-nucleotide exchange protein (GEP) activity, and the PH domain interacts with phospholipids and is responsible for association of PSCDs with membranes. Members of this family appear to mediate the regulation of protein sorting and membrane trafficking. The PSCD4 exhibits GEP activity in vitro with both ARF1 and ARF5 but is inactive with ARF6. The PSCD4 and PSCD1 gene structures are very similar (Entrez database).


Solute carrier family 22 (organic cation transporter) member 1. Polyspecific organic cation transporters in the liver, kidney, intestine, and other organs are critical for elimination of many endogenous small organic cations as well as a wide array of drugs and environmental toxins. Solute carrier family 22 (organic cation transporter) member 1 (SLC22A1 alias OCT1) is one of three similar cation transporter genes located in a cluster on chromosome 6. The encoded protein contains twelve putative transmembrane domains and is a plasma integral membrane protein. Two transcript variants encoding two different isoforms have been found for this gene, but only the longer variant encodes a functional transporter (Entrez database). OCT1 is the main organic cation uptake system in hepatocytes and has common features with organic cation uptake over the basolateral membrane of renal proximal tubules (Grundemann et al., 1994). Together with other 24 membrane transporters OCT1 have potential roles in drug response, as determined by phylogenetic and population-genetics analysis in a large population (Leabman et al., 2003). Changes at evolutionarily conserved positions of OCT1 are strong predictors of decreased function and suggested that a combination of evolutionary conservation and chemical change might be a stronger predictor of function (Shu et al., 2003). hOCT1, which is expressed in the intestine and liver, is likely to play a major role in the intestinal absorption and hepatic disposition of ranitidine and famotidine in humans (Bourdet et al., 2005). OCT1 and OCT2 mediate luminal Ach (acetylcholine) release in human airways and suggest that ACh release is blocked after inhalation of budesonide (Lips et al., 2005). The expression of organic cation transporters rOCT1 and rOCT2 is reduced in experimental diabetes in rats (Grover et al., 2004). When mice were given metformin, the blood lactate concentration significantly increased in the wild-type mice, whereas only a slight increase was observed in Oct1(−/−) mice. The plasma concentration of metformin exhibited similar time profiles between the wild-type and Oct1(−/−) mice, suggesting that the liver is the key organ responsible for the lactic acidosis and the Oct1 gene is involved in lactic acidosis caused by metformin (Wang et al., 2003).


Tyrosine-protein kinase-like 7 precursor. Tyrosine-protein kinase-like 7 precursor (PTK7 alias CCK4 (Colon carcinoma kinase 4, CCK-4) is a receptor protein tyrosine kinase which transduce extracellular signals across the cell membrane. A subgroup of these kinases lack detectable catalytic tyrosine kinase activity but retain roles in signal transduction. The protein encoded by this gene is a member of this subgroup of tyrosine kinases and may function as a cell adhesion molecule. This gene is thought to be expressed in colon carcinomas but not in normal colon, and therefore may be a marker for or may be involved in tumor progression. Five transcript variants encoding five different isoforms have been found for this gene (Entrez database).


Cytidine monophosphate-N-acetylneuraminic acid hydroxylase. Cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) is also known as CMP-N-acetylneuraminate monooxygenase, CMP-NeuAc hydroxylase, CMP-Neu5Ac hydroxylase, CMP-sialic acid hydroxylase (CSAH), CMP-N-acetylneuraminic acid hydroxylase). Sialic acids are terminal components of the carbohydrate chains of glycoconjugates involved in ligand-receptor, cell-cell, and cell-pathogen interactions. The two most common forms of sialic acid found in mammalian cells are N-acetylneuraminic acid (Neu5Ac) and its hydroxylated derivative, N-glycolylneuraminic acid (Neu5Gc). Studies of sialic acid distribution show that Neu5Gc is not detectable in normal human tissues although it was an abundant sialic acid in other mammals. Neu5Gc is, in actuality, immunogenic in humans. The absense of Neu5Gc in humans is due to a deletion within the human gene CMAH encoding cytidine monophosphate-N-acetylneuraminic acid hydroxylase, an enzyme responsible for Neu5Gc biosynthesis. Sequences encoding the mouse, pig, and chimpanzee hydroxylase enzymes were obtained by cDNA cloning and found to be highly homologous. However, the homologous human cDNA differs from these cDNAs by a 92-bp deletion in the 5′ region. This deletion, corresponding to exon 6 of the mouse hydroxylase gene, causes a frameshift mutation and premature termination of the polypeptide chain in human. It seems unlikely that the truncated human hydroxylase mRNA encodes for an active enzyme explaining why Neu5Gc is undetectable in normal human tissues. Human genomic DNA also shows evidence of this deletion which does not occur in the genomes of African great apes. Nonetheless, the CMAH gene maps to 6p21.32 in humans and great apes indicating that mutation of the CMAH gene occurred following human divergence from chimpanzees and bonobos (Entrez database). Studies indicate that the CMAH gene is inactivated shortly before the time when brain expansion began in humankind's ancestry, approximately 2.1-2.2 mya. In this regard, it is of interest that although Neu5Gc is the major sialic acid in most organs of the chimpanzee, its expression is selectively down-regulated in the brain (Chou et al, 2002).


Solute carrier family 24 (sodium/potassium/calcium exchanger), member 3. Solute carrier family 24 (sodium/potassium/calcium exchanger) member 3 is also known as SLC24A3 or NCKX3. The majority of TM neurones express NCX1, NCX2 and NCKX3 (Sergeeva et al, 2003). The N-terminal region of NCKX3, although not essential for expression, increased functional activity at least 10-fold and may represent a cleavable signal sequence. NCKX3 transcripts were most abundant in brain, with highest levels found in selected thalamic nuclei, in hippocampal CA1 neurons, and in layer IV of the cerebral cortex. Many other tissues also expressed NCKX3 at lower levels, especially aorta, uterus, and intestine, which are rich in smooth muscle (Kraev et al, 2001).


Somatostatin receptor type 3. Somatostatin receptor type 3 is also known as SS3R, SSR-28 or SSTR3. Somatostatin acts at many sites to inhibit the release of many hormones and other secretory proteins. The biological effects of somatostatin are probably mediated by a family of G protein-coupled receptors that are expressed in a tissue-specific manner. SSTR3 is a member of the superfamily of receptors having seven transmembrane segments and is expressed in highest levels in brain and pancreatic islets. SSTR3 is functionally coupled to adenylyl cyclase (Entrez database). Five SSTR subtypes are variably expressed at the mRNA level in breast tumors with 91% of samples showing SSTR1, 98% SSTR2, 96% SSTR3, 76% SSTR4, and 54% SSTR5. Levels of SSTR mRNA, when corrected for beta-actin levels, were highest for SSTR3 (Kumar et al, 2005). All five SSTRs were differentially expressed as membrane and cytoplasmic proteins in cortical neurons with significant variations in control vs. alzheimer diseased (AD) brain. In AD cortical brain region, somatostatin and neuropeptide-Y-positive neurons decreased (>70%). SSTR3 was the only receptor subtype that increased in AD cortex (Kumar, 2005). SSTR3 is expressed in the HCC cells, but not in the L-02 cells, which suggests a molecular basis for the HCC-selective effects of octreotide (Liu et al, 2004). SSTR3 protein existed in the membrane of gastric cancer cells. In normal gastric mucosa, SSTR3 protein distributed to the cellular membrane and cytoplasm or interstitial tissue in submucosa. The expression of SSTR3 protein was significantly lower in gastric cancer compared with normal mucosa. Moreover, the poor-differentiated adenocarcinoma was lower than the well-differentiated adenocarcinoma, and the similar result in cell lines. Octreotide could inhibit the growth and induce the apoptosis of gastric cancer and normal epithelial cells that expressed SSTR3, but didn't affect the cells with no or weakly expression of SSTR3 (Hu et al, 2004). SSTR3 mRNA is confined to the pituitary, hypothalamus, and spinal cord from early to midgestation (Goodyer et al, 2004). In vitro, octreotide inhibited the proliferation, invasion, and differentiation of HUVECs elicited by VEGF. RT-PCR analysis demonstrated that HUVECs expressed the somatostatin receptor subtype SSTR3. In vivo, octreotide was sufficiently potent to suppress nude mice corneal neovascularization induced by tumor tissues from LCI-D20 (Jia et al, 2003). In renal cell tumors, SSTR3 transcripts were completely absent. In breast cancer tissue, SSTR subtypes were transcribed independently of patient age, menstrual status, diagnosis, histological grade, and levels of estrogen receptor and progesterone receptor (Vikic-Topic et al, 1995).


Bone morphogenetic protein 7 precursor. Bone morphogenetic protein 7 precursor (BMP-7) is also known as Osteogenic protein 1 (OP-1) or Eptotermin alfa. The bone morphogenetic proteins (BMPs) are a family of secreted signaling molecules that can induce ectopic bone growth. Many BMPs are part of the transforming growth factor-beta (TGFB) superfamily. BMPs were originally identified by an ability of demineralized bone extract to induce endochondral osteogenesis in vivo in an extraskeletal site. Based on its expression early in embryogenesis, the BMP encoded by this gene has a proposed role in early development. In addition, the fact that this BMP is closely related to BMP5 and BMP7 has lead to speculation of possible bone inductive activity (Entrez database).


Caspase recruitment domain protein 10. Caspase recruitment domain protein 10 (CARD10) is also known as CARD-containing MAGUK protein 3 (CARMA 3). The caspase recruitment domain (CARD) is a protein module that consists of 6 or 7 antiparallel alpha helices. It participates in apoptosis signaling through highly specific protein-protein homophilic interactions. CARDs induce nuclear factor kappa-B (NFKB; MIM 164011) activity through the IKK (e.g., IKBKB; MIM 603258) complex. CARD9 (MIM 607212), CARD10, CARD11 (MIM 607210), and CARD14 (MIM 607211) interact with BCL10 (MIM 603517) and are involved in NFKB signaling complexes. Except for CARD9, these CARD proteins are members of the membrane-associated guanylate kinase (MAGUK) family (OMIM database). CARMA3 physically associate with Ikappa kinase gamma/NFkappaB essential modulator (IkappaKgamma-NEMO) in lymphoid and non-lymphoid cells. Expression of the NEMO-binding region of CARMA3 exerts a dominant negative effect on BCL10-mediated activation of NfkappaB (Stilo et al, 2004). CARD10 is a novel BCL10 interactor that belongs to the membrane-associated guanylate kinase family, a class of proteins that function to organize signaling complexes at plasma membranes. When expressed in cells, CARD10 binds to BCL10 and signals the activation of NF-kappaB through its N-terminal effector CARD domain. It is proposed that CARD10 functions as a molecular scaffold for the assembly of a BCL10 signaling complex that activates NF-kappaB (Wang et al, 2001).


Neutrophil cytosol factor 4. Neutrophil cytosol factor 4 (NCF-4), also known as Neutrophil NADPH oxidase factor 4 or p40-phox, is a cytosolic regulatory component of the superoxide-producing phagocyte NADPH-oxidase, a multicomponent enzyme system important for host defense. This protein is preferentially expressed in cells of myeloid lineage. It interacts primarily with neutrophil cytosolic factor 2 (NCF2/p67-phox) to form a complex with neutrophil cytosolic factor 1 (NCF1/p47-phox), which further interacts with the small G protein RAC1 and translocates to the membrane upon cell stimulation. This complex then activates flavocytochrome b, the membrane-integrated catalytic core of the enzyme system. The PX domain of this protein can bind phospholipid products of the PI(3) kinase, which suggests its role in PI(3) kinase-mediated signaling events. The phosphorylation of this protein was found to negatively regulate the enzyme activity. Alternatively spliced transcript variants encoding distinct isoforms have been observed (Entrez database).


Cadherin EGF LAG seven-pass G-type receptor 1 precursor. Cadherin EGF LAG seven-pass G-type receptor 1 precursor (CELSR1, ME2, CDHF9) also known as Flamingo homolog 2 (HFM12, FM12). is a member of the flamingo subfamily, part of the cadherin superfamily. The flamingo subfamily consists of nonclassic-type cadherins; a subpopulation that does not interact with catenins. The flamingo cadherins are located at the plasma membrane and have nine cadherin domains, seven epidermal growth factor-like repeats and two laminin A G-type repeats in their ectodomain. They also have seven transmembrane domains, a characteristic unique to this subfamily. It is postulated that these proteins are receptors involved in contact-mediated communication, with cadherin domains acting as homophilic binding regions and the EGF-like domains involved in cell adhesion and receptor-ligand interactions. This particular member is a developmentally regulated, neural-specific gene which plays an unspecified role in early embryogenesis (Entrez database). Expression of the CELSR/Flamingo homologue, c-fmi1, in the early avian embryo indicates a conserved role in neural tube closure and additional roles in asymmetry and somitogenesis (Formstone and Mason, 2005). Each CELSR is expressed prominently in the developing brain following a specific pattern, suggesting that they serve distinct functions (Tissir et al, 2002).


Platelet-derived growth factor B chain precursor. Platelet-derived growth factor B chain precursor (PDGF B-chain, PDGFB, SIS, SSV, PDGF2, c-sis) is a member of the platelet-derived growth factor family. The four members of this family are mitogenic factors for cells of mesenchymal origin and are characterized by a motif of eight cysteines. This gene product can exist either as a homodimer (PDGF-BB) or as a heterodimer with the platelet-derived growth factor alpha polypeptide (PDGF-AB), where the dimers are connected by disulfide bonds. Mutations in this gene are associated with meningioma. Reciprocal translocations between chromosomes 22 and 7, at sites where this gene and that for COL1A1 are located, are associated with a particular type of skin tumor called dermatofibrosarcoma protuberans resulting from unregulated expression of growth factor. Two splice variants have been identified for this gene (Entrez database). Medulloblastomas contained the highest amounts of PDGF B-chain, some four to eight times more than that in control brain tissue. Tumors that contained a high level of PDGF B-chain showed high proliferative activity (Nakamura et al, 1993). Trichostatin A (TSA) activates reporter gene constructs driven by the human platelet-derived growth factor B (PDGF-B) gene promoter. This activation showed an inverse correlation with the cell type-specific transcriptional activities of the promoter (Ulleras et al, 2001). Tissue specificity was not clear in the 5′ upstream region alone, and regulation by gene methylation or by elements other than in the 5′ region seemed to be necessary (Takimoto et al, 1993). Platelet-derived growth factor B-chain was also abnormally methylated in 4 of 13 (31%) multinodular goiters (MNG), 17 of 24 (71%) follicular adenomas (FA), and 9 of 13 (69%) papillary carcinomas (PC) (Matsuo et al, 1993). Gene expression of PDGF-A and PDGF-B mRNA were increased 22- and 6-fold, respectively, in biopsies from patients with diabetic nephropathy compared with control tissue (Langham et al, 2003). Adult SPC-PDGFB transgenic mice exhibited lung pathology characterized by enlarged airspaces, inflammation, and fibrosis (Hoyle et al, 1999). The transition between hyperplasia (complete hydatidiform mole) and neoplasia (choriocarcinoma) in these cells correlates with at least a 10- to 20-fold activation of the PDGF-B gene (Holmgren et al, 1993).


Transmembrane protease serine 6. Transmembrane protease serine 6 (TMPRSS6) is a type II transmembrane serine proteinase that is found attached to the cell surface. The encoded protein may be involved in matrix remodeling processes in the liver (Entrez database).


Bcl-2 interacting killer. Bcl-2 interacting killer (BIK, BP4, BIP1, BBC1) also known as Apoptosis inducer NBK (NBK) is known to interact with cellular and viral survival-promoting proteins, such as BCL2 and the Epstein-Barr virus in order to enhance programmed cell death. Because its activity is suppressed in the presence of survival-promoting proteins, this protein is suggested as a likely target for antiapoptotic proteins. This protein shares a critical BH3 domain with other death-promoting proteins, BAX and BAK (Entrez database). Human blk is expressed only in B lymphocytes (Drebin et al, 1995). The two human blk RNAs arise from the transcription of the blk gene by two distinct promoters and that these promoters may be subject to regulation by different trans-acting factors (Lin et al, 1995). P55blk and p53/p56lyn may be particularly good candidates for the membrane immunoglobulin-activated tyrosine kinase (Law et al, 1992). There is a role of blk kinase in anti-mu-mediated pathway to cell cycle arrest (Yao et al, 1993). Blk is down-regulated in a clinically distinct aggressive subset of B-CLL completely resistant in vitro to irradiation-induced apoptosis (Vallat et al, 2003). Despite the absence of Blk, the development, in vitro activation, and humoral immune responses of B cells to T-cell-dependent and -independent antigens are unaltered. These data are consistent with functional redundancy of Blk in B-cell development and immune responses (Texido et al, 2000). Expression of constitutively active Blk in the T lineage resulted in the appearance of clonal, thymic lymphomas composed of intermediate single positive cells (Malek et al, 1998).


PKHD1. PKHD1 stands for polycystic kidney and hepatic disease 1 (autosomal recessive) and is also known as FCYT, ARPKD, TIGM1. The protein encoded by this gene is predicted to have a single transmembrane (TM)-spanning domain and multiple copies of an immunoglobulin-like plexin-transcription-factor domain. Alternative splicing results in two transcript variants encoding different isoforms. Other alternatively spliced transcripts have been described, but the full length sequences have not been determined. Several of these transcripts are predicted to encode truncated products which lack the TM and may be secreted. Mutations in this gene cause autosomal recessive polycystic kidney disease, also known as polycystic kidney and hepatic disease-1 (Entrez database). HNF-1beta mutant mice show decreased expression of Pkhd1, the gene that is mutated in humans with autosomal-recessive polycystic kidney disease (ARPKD) (Igarashi et al, 2005). A total of 263 different PKHD1 mutations (639 mutated alleles) are included in the locus-specific database. Except for a few population-specific founder alleles and the common c.107C>T (p.Thr36Met) missense change, PKHD1 is characterized by significant allelic diversity (Bergmann et al, 2005). Polyductin is part of the group of polycystic kidney disease (PKD)-related proteins expressed in primary apical cilia. It probably serves in other subcellular functional roles. The detection of three different products using two antisera, with evidence for distinct subcellular localizations, suggests that PKHD1 encodes membrane-bound and soluble isoforms (Menezes et al, 2004). Renal cyst formation is accompanied by a drastic defect in the transcriptional activation of Umod, Pkhd1 and Pkd2 genes, whose mutations are responsible for distinct cystic kidney syndromes. In vivo chromatin immunoprecipitation experiments demonstrated that HNF1beta binds to several DNA elements in murine Umod, Pkhd1, Pkd2 and Tg737/Polaris genomic sequences (Gresh et al 2004). During embryogenesis, PKHD1 is widely expressed in epithelial derivatives, including neural tubules, gut, pulmonary bronchi, and hepatic cells. In the kidneys of the pck rats, the rat model of which is genetically homologous to human ARPKD, the level of PKHD1 was significantly reduced but not completely absent. In cultured renal cells, the PKHD1 gene product colocalized with polycystin-2, the gene product of autosomal dominant polycystic disease type 2, at the basal bodies of primary cilia (Zhang et al, 2004). PKHD1 was identified to be mutated in ARPKD (Onuchic et al, 2002).


Myosin-18B. Myosin-18B, also known as Myosin XVIIIb alias BK125H2.1, may regulate muscle-specific genes when in the nucleus and may influence intracellular trafficking when in the cytoplasm. The encoded protein functions as a homodimer and may interact with F actin. Mutations in this gene are associated with lung cancer (Entrez database). Genetic and epigenetic alterations of the MYO18B gene was analyzed in colorectal cancers. Alleic imbalance at the MYO18B locus was detected in 16 of 43 (40%) informative cases. Mutations of the MYO18B gene were detected in 2 of 11 (18%) cell lines and 1 of 47 (2%) surgical specimens. Nine of 11 (82%) cell lines showed reduced MYO18B expression, which was restored in all 9 by treatment with 5-aza-2′-deoxycytidine and/or trichostatin A (TSA). Although hypermethylation of the promoter CpG island for MYO18B was not detected, a significant correlation was observed between the level of MYO18B expression and the level of acetylation of histones H3 and H4 in 6 cell lines with and without TSA treatment (Nakano et al, 2005). Missense MYO18B mutations were detected in 1 of 4 (25%) ovarian cancer cell lines and in 1 of 17 (5.9%) primary ovarian cancers. MYO18B expression was reduced in all 4 ovarian cancer cell lines and in 12 of 17 (71%) of primary ovarian cancers. MYO18B expression was restored by treatment with 5-aza-2′-deoxycytidine and/or trichostatin A in 3 of 4 cell lines with reduced MYO18B expression, and hypermethylation of the promoter CpG island for MYO18B was observed in 2 of these 3 cell lines. Its hypermethylation was also observed in 2 of 15 (13%) primary ovarian cancers (Yanaihara et al, 2004). MYO18B, located at chromosome 22q12.1 and found that it is frequently deleted, mutated, and hypermethylated in lung cancers (Nishioka et al, 2002).


GAS2-likeprotein 1. GAS2-like protein 1 (GAS2L1) is also known as Growth arrest-specific 2-like 1 or GAS2-related protein on chromosome 22 (GAR22 protein, GAR22). The protein encoded by this gene, a member of the GAS2 family, is similar in sequence to the mouse protein Gas2, an actin-associated protein expressed at high levels in growth-arrested cells. Expression of the mouse Gas2 gene is negatively regulated by serum and growth factors. Three transcript variants encoding two different isoforms have been found for this gene (Entrez database). Although hGAR22 and mGAR22 mRNAs are expressed nearly ubiquitously, mGAR22 protein can only be detected in testis and brain.


Furthermore, only the beta isoform is present in these tissues. GAR22beta expression is induced in a variety of cultured cells by growth arrest. The absolute amounts of GAR22beta protein expressed are low (Goriounov et al, 2003). The regulation of Gas2 biosynthesis reflects the pattern of mRNA expression: its relative level is tightly associated with growth arrest. Gas2 seems to be regulated also at the posttranslational level via a phosphorylation mechanism (Brancolini et al, 1992).


Ras and Rab interactor 2. Ras and Rab interactor 2 (RIN2, RASSF4) The RAB5 protein is a small GTPase involved in membrane trafficking in the early endocytic pathway. The protein encoded by this gene binds the GTP-bound form of the RAB5 protein preferentially over the GDP-bound form, and functions as a guanine nucleotide exchange factor for RAB5. The encoded protein is found primarily as a tetramer in the cytoplasm and does not bind other members of the RAB family (Entrez database). RASSF4 (AD037) shows approximately 25% identity with RASSF1A and 60% identity with RASSF2. RASSF4 binds directly to activated K-Ras in a GTP-dependent manner via the effector domain, thus exhibiting the basic properties of a Ras effector. Overexpression of RASSF4 induces Ras-dependent apoptosis in 293-T cells and inhibits the growth of human tumor cell lines. Although broadly expressed in normal tissue, RASSF4 is frequently down-regulated by promoter methylation in human tumor cells. Thus, RASSF4 appears to be a new member of the RASSF family of potential Ras effector/tumor suppressors (Eckfeld et al, 2004). It was demonstrated that the expression of RASSF4/AD037 was lost in 12.5% (1/8) of NPC cell lines/xenografts. Bisulfite sequencing analysis revealed dense methylation in the promoter region of RASSF4/AD037 in the cell line. Restoration of RASSF4/AD037 mRNA was observed by treatment with a demethylating agent (Chow et al, 2004).


Forkhead box C1. Forkhead box C1 (FOXC1, FKHL7, IRID1, FREAC3, ARA, IGDA, IHG1) belongs to the forkhead family of transcription factors which is characterized by a distinct DNA-binding forkhead domain. The specific function of this gene has not yet been determined; however, it has been shown to play a role in the regulation of embryonic and ocular development. Mutations in this gene cause various glaucoma phenotypes including primary congenital glaucoma, autosomal dominant iridogoniodysgenesis anomaly, and Axenfeld-Rieger anomaly (Entrez database).


GRB2-related adaptor protein 2. GRB2-related adaptor protein 2 (GRAP2, Grf40, GrbX, GRBLG, GADS, Mona, P38; GRID; GRPL; GRB2L; GRAP-2) a member of the GRB2/Sem5/Drk family. This member is an adaptor-like protein involved in leukocyte-specific protein-tyrosine kinase signaling. Like its related family member, GRB2-related adaptor protein (GRAP), this protein contains an SH2 domain flanked by two SH3 domains. This protein interacts with other proteins, such as GRB2-associated binding protein 1 (GAB1) and the SLP-76 leukocyte protein (LCP2), through its SH3 domains. Transcript variants utilizing alternative polyA sites exist (Entrez database). GRAP2 and GPR51, were found to respond to low-dose radiation but not to high-dose radiation in G1-arrested normal human skin fibroblasts (Ding et al, 2005). Gads adaptor protein is expressed in many hematopoietic tissues, including bone marrow, lymph node, and spleen. Using intracellular staining, Gads protein was detected in a number of cells, including B cells, T cells, NK cells, monocytes, and plasmacytoid DC, but not in macrophages, neutrophils, or monocyte-derived DC. Gads may have a negative regulatory role in signaling through survival pathways, and is necessary for normal homeostatic proliferation in B cells (Yankee et al, 2005). GRAP genes were up-regulated in salivary glands of the MRL/lpr (an Sjogren's syndrome (SS) mouse model) (Shiraiwa et al, 2004). −2000 to +150 genomic region relative to the Mona gene transcription start site is sufficient to direct specific reporter gene expression in T cell lines, Jurkat, and MOLT-4 and in the immature myeloid cell lines, KG1a and RC2A. Deletion analysis and electrophoresis mobility shift assay identified several cis regulatory elements: overlapping initiator sequences, one interferon response factor-2 (IRF-2)-binding site at position −154, one GC box recognized by Sp1 and Sp3 at position −52, and two acute myeloid leukemia (AML)-1 binding sites at positions −70 and −13 (Guyot and Mouchiroud, 2003). It was reported that a lineage-restricted transcription of the Mona gene is controlled by specific promoters (Guyot et al, 2002).


RASSF2. RASSF2 stands for Ras association (RalGDS/AF-6) domain family 2 and is also known as KIAA0168 or DKFZp781O1747. This gene encodes a protein that contains a Ras association domain. Similar to its cattle and sheep counterparts, this gene is located near the prion gene. The specific function of this gene has not yet been determined. Three alternatively spliced transcript variants of this gene encoding two distinct isoforms have been reported (Entrez database). Hypermethylation of RASSF2 in at least one of the regions examined was detected in seven (70%) of the 10 gastric cancer cell lines; two (20%) exhibited hypermethylation in all the regions examined including the transcription start site and lost expression of RASSF2 mRNA, which could, however, be restored by 5-aza-2′ deoxycytidine treatment, while the other five (50%) cell lines exhibited hypermethylation at the 5′- and/or 3′-edge, with four of them expressing RASSF2 mRNA. In primary gastric cancers and corresponding non-neoplastic gastric epithelia, frequencies of RASSF2 methylation ranged from 29% (23 out of 78) to 79% (62 out of 78) and 3% (two out of 78) to 60% (47 out of 78), respectively, at different CpG sites examined (Endoh et al, 2005). Aberrant methylation and histone deacetylation of RASSF2 was associated with the gene's silencing in CRC. The activities of RASSF2, which were distinct from those of RASSF1, included induction of morphologic changes and apoptosis; moreover, its ability to prevent cell transformation suggests that RASSF2 acts as a tumor suppressor in CRC. Primary CRCs that showed K-ras/BRAF mutations also frequently showed RASSF2 methylation, and inactivation of RASSF2 enhanced K-ras-induced oncogenic transformation. RASSF2 methylation was also frequently identified in colorectal adenomas (Akino et al, 2005). RASSF2A is frequently inactivated in CRCs by CpG island promoter hypermethylation, and that epigenetic (RASSF2A) and genetic (K-ras) changes are mutually exclusive and provide alternative pathways for affecting Ras signalling (Hesson et al, 2005). RASSF2 binds directly to K-Ras in a GTP-dependent manner via the Ras effector domain. However, RASSF2 only weakly interacts with H-Ras. Moreover, RASSF2 promotes apoptosis and cell cycle arrest and is frequently down-regulated in lung tumor cell lines (Vos et al, 2003).


Glutamate receptor, ionotropic kainate 2 precursor. Glutamate receptor, ionotropic kainate 2 precursor (GRIK2 RP3-438O4.2 (Vega gene ID)) is also known as Glutamate receptor 6 (GluR-6) or Excitatory amino acid receptor 4 (EAA4). This gene encodes a subunit of a kainate glutamate receptor. Glutamate receptors mediate the majority of excitatory neurotransmission in the brain. This receptor may have a role in synaptic plasticity and may be important for learning and memory. It also may be involved in the transmission of light information from the retina to the hypothalamus. The structure and function of the encoded protein is changed by RNA editing. Alternatively spliced transcript variants encoding distinct isoforms have been described for this gene (Entrez database). Histone methylation marks were studied at proximal promoters of 16 ionotropic and metabotropic glutamate receptor genes (GRIN1,2A-D; GRIA1,3,4; GRIK2,4,5; GRM1,3,4,6,7) in cerebellar cortex collected across a wide age range from midgestation to 90 years old. Levels of di- and trimethylated histone H3-lysine 4, which are associated with open chromatin and transcription, showed significant differences between promoters and a robust correlation with corresponding mRNA levels in immature and mature cerebellar cortex. In contrast, levels of trimethylated H3-lysine 27 and H4-lysine 20, two histone modifications defining silenced or condensed chromatin, did not correlate with transcription but were up-regulated overall in adult cerebellum (Stadler et al, 2005). Maternal transmission disequilibrium of GRIK2 was observed with a significance of p=0.03 (Bah et al, 2004). Deletion data singled out GRIK2 as the gene most frequently affected by deletions of 6q in acute lymphocytic leukemia (ALL). Sequence analysis of GRIK2 in 14 ALL cases carrying heterozygous 6q deletions revealed a constitutional and paternally inherited C to G substitution in exon 6 encoding for an amino acid change in one patient. The substitution was absent among 232 normal alleles tested, leaving open the possibility that heterozygous carriers of such mutations may be susceptible to ALL. Although low in all normal hematopoietic tissues, quantitative reverse transcription-PCR showed higher baseline GRIK2 expression in thymus and T cells than other lineages. Among T-cell ALL patients, 6q deletion was associated with a statistically significant reduction in GRIK2 expression (P=0.0001). By contrast, elevated GRIK2 expression was measured in the myelomonocytic line THP-1 and in one patient with common ALL. Finally, significant levels of GRIK2 expression were detected in prostate, kidney, trachea, and lung, raising the possibility that this gene may be protective against multiple tumor types (Sinclair et al, 2004).


T-box transcription factor TBX1. T-box transcription factor TBX1 is also known as T-box protein 1, or Testis-specific T-box protein (TBX1, DGS, TGA, CAFS, CTHM, DGCR, DORV, VCFS, TBX1C). This gene is a member of a phylogenetically conserved family of genes that share a common DNA-binding domain, the T-box. T-box genes encode transcription factors involved in the regulation of developmental processes. This gene product shares 98% amino acid sequence identity with the mouse ortholog. DiGeorge syndrome (DGS)/velocardiofacial syndrome (VCFS), a common congenital disorder characterized by neural-crest-related developmental defects, has been associated with deletions of chromosome 22q11.2, where this gene has been mapped. Studies using mouse models of DiGeorge syndrome suggest a major role for this gene in the molecular etiology of DGS/VCFS. Several alternatively spliced transcript variants encoding different isoforms have been described for this gene (Entrez database). Association was demonstrated between variants and haplotypes of remaining TBX1 gene and manifestations of congenital heart defects in 22q11.2 deletion patients (Rauch et al, 2004). Tbx1 haploinsufficiency causes aortic arch abnormalities in mice because of early growth and remodeling defects of the fourth pharyngeal arch arteries (Vitelli et al, 2002). Tbx1 may trigger signals from the pharyngeal endoderm directed to the underlying mesenchyme. Expression patterns of Fgf8 and Fgf10, which partially overlap with Tbx1 expression pattern, are altered in Tbx1 (−/−) mutants (Vitelli et al, 2002). RA could produce an altered Tbx1 expression pattern in zebrafish. In addition, RA could repress Tbx1 expression in a dose-dependant manner (Zhang et al, 2006).


Cadherin-like 22. Cadherin-like 22 (CDH22, C20orf25 dJ998H6.1 MGC39564 is a member of the cadherin superfamily. The gene product is composed of five cadherin repeat domains and a cytoplasmic tail similar to the highly conserved cytoplasmic region of classical cadherins. Expressed predominantly in the brain, this putative calcium-dependent cell adhesion protein may play an important role in morphogenesis and tissue formation in neural and non-neural cells during development and maintenance of the brain and neuroendocrine organs (Entez database).


Nuclear factor of activated T-cells cytoplasmic 2. Nuclear factor of activated T-cells cytoplasmic 2 (NFATC2) is also known as T cell transcription factor NFAT1 or NFAT pre-existing subunit (NF-ATP, NFATp). This gene is a member of the nuclear factor of activated T cells (NFAT) family. The product of this gene is a DNA-binding protein with a REL-homology region (RHR) and an NFAT-homology region (NHR). This protein is present in the cytosol and only translocates to the nucleus upon T cell receptor (TCR) stimulation, where it becomes a member of the nuclear factors of activated T cells transcription complex. This complex plays a central role in inducing gene transcription during the immune response. Alternate transcriptional splice variants, encoding different isoforms, have been characterized (Entrez database). The expression levels of the NFAT family members NFAT1, -2, and -4 were normal in the SCID patients' T cells, dephosphorylation and nuclear translocation of these NFAT proteins occurred very transiently and incompletely upon stimulation (Feske et al, 2000). Two of the seven kinase inhibitors, staurosporine (St) and bisindolylmaleimide I (BI), resulted in the dephosphorylation and nuclear localization of NFATp. Treatment of cells with ionomycin resulted in NFATp dephosphorylation and nuclear localization (Feske et al, 2000). NFAT1 mRNA is preferentially expressed in mature CD4(+) single-positive cells (Amasaki et al, 2000). Continued culture in the presence of polarizing cytokines established a selective pattern of histone acetylation on both cytokine genes. This correlated with restricted access of the transcription factor NFAT1 to these gene regulatory regions as well as mutually exclusive gene expression by the differentiated T cells (Avni et al, 2001). The level of NFATc2 binding to NFAT motifs in the CD3gamma gene promoter was greatly increased in the abnormal T cells from hypereosinophilic syndrome (Willard-Gallo et al, 2005).


MyoD family inhibitor. MyoD family inhibitor (MDFI, I-MF) is also known as Myogenic repressor I-mf. It is a transcription factor that negatively regulates other myogenic family proteins. Studies of the mouse homolog, 1-mf, show that it interfers with myogenic factor function by masking nuclear localization signals and preventing DNA binding. Knockout mouse studies show defects in the formation of vertebrae and ribs that also involve cartilage formation in these structures (Entrez database). I-mfa domain proteins interact with the Axin complex and affect Axin regulation of both the Wnt and the JNK activation pathways (Kusano S and Raab-Traub, 2002). I-mfa is expressed at a low level in an osteoblast-like cell line, MC3T3E1, and a pluripotent differentiation modulator, 1,25-dihydroxyvitamin D(3), specifically enhanced 1-mfa mRNA expression (Tsuji et al, 2001). I-mfa plays an important role in trophoblast and chondrogenic differentiation by negatively regulating a subset of lineage-restricted bHLH proteins (Kraut et al, 1998).


TGM3. TGM3 alias TGE, MGC126249 or MGC126250 stands for transglutaminase 3 (E polypeptide, protein-glutamine-gamma-glutamyltransferase). Transglutaminases are enzymes that catalyze the crosslinking of proteins by epsilon-gamma glutamyl lysine isopeptide bonds. While the primary structure of transglutaminases is not conserved, they all have the same amino acid sequence at their active sites and their activity is calcium-dependent. The protein encoded by this gene consists of two polypeptide chains activated from a single precursor protein by proteolysis. The encoded protein is involved the later stages of cell envelope formation in the epidermis and hair follicle (Entrez database). Significantly higher levels of keratin (Ker)-14 and -17 mRNAs, combined with lower levels of Ker-4, Ker-13 and transglutaminase 3 (TG-3) transcripts, were observed in OSCC (Oral Squamous Cell Carcinoma) and severely dysplastic tissues, whereas this expression profile was reversed in hyperplasia and in mild to moderate dysplasia (Ohkura et al, 2005). TGM3 plays a important role in the epidermis differentiation in embryogenesis (Zhang et al, 2005). Transglutaminase 1, 2, and 3 could be involved in cross-linking of huntingtin into intranuclear inclusions in HD (Huntington disease). It was suggested that inhibiting transglutaminase should be explored as a potential treatment strategy for HD (Zainelli et al, 2005). Immunostaining for transglutaminase3 was absent or faint throughout almost the entire suprabasal epidermis in NTS (Netherton Syndrome) (Raghunath et al, 2004). It was revealed that genes involved in squamous cell differentiation were coordinately downregulated, including annexin I, small proline-rich proteins (SPRRs), calcium-binding S100 proteins (S100A8, S100A9), transglutaminase (TGM3), cytokeratins (KRT4, KRT13), gut-enriched Krupple-like factor (GKLF) and cystatin A, in human esophageal squamous cell carcinoma (ESCC) (Luo et al, 2004). TGM3 is downregulated in head and neck squamous cell carcinoma (Gonzalez et al, 2003). Immunohistochemical analysis of the skin revealed that the enzyme is present in the cells of the granular and cornified layers consistent with its role in cornified envelope formation. In cultured keratinocytes, TGase 3 was expressed in differentiating cells coincident with profilaggrin and keratin 10 expressions (Hitomi et al, 2003).


Disheveled associated activator of morphogenesis 2. Disheveled associated activator of morphogenesis 2 (DAAM2, KIAA0381, MGC90515, dJ90A20A.1, RP1-278E11.1) regulates the morphogenetic movements of vertebrate gastrulation in a Wnt-dependent manner through direct interactions with Dsh/Dvl and RhoA (Nakaya et al, 2004). The observed expression patterns in developing central nervous tissues suggested that vertebrate Daam genes were involved in pivotal steps in neuronal cell differentiation and movement (Kida et al, 2004).


OTTHUMG00000030521, AC000095.4. OTTHUMG00000030521, AC000095.4 (Vega gene ID) alias Em:AC000095.C22.4 putative processed transcript is probably DiGeorge Syndrome gene B (Vega gene Report).


CAP-binding protein complex interacting protein 1 isoform. CAP-binding protein complex interacting protein 1 isoform (a Source: RefSeq_peptide NP073622) alias FLJ23588; DJBP; HSCBCIP1; KIAA1672; dJ185D5.1). DJBP mRNA was found to be specifically expressed in the testis. In addition to the binding of DJBP to the COOH-terminal region of DJ-1, DJBP was also found to bind in vitro and in vivo to the DNA-binding domain of the androgen receptor (AR) in a testosterone-dependent manner and to be colocalized with DJ-1 or AR in the nucleus. Furthermore, a co-immunoprecipitation assay showed that the formation of a ternary complex between DJ-1, DJBP, and AR occurred in cells in which DJ-1 bound to the AR via DJBP. It was found that DJBP repressed a testosterone-dependent AR transactivation activity in monkey Cos1 cells by recruiting histone deacetylase (HDAC) complex (Niki et al, 2003). Necropsy tissues from 11 cases were analyzed with 1 tumor specimen found to have HIV integrated in chromosome 22q13.2 and within 300 kb of HSCBCIP1 (CAP-binding protein complex interacting homologue). Tumor-specific primers were then used to screen uninvolved tissue from the same patient, which did not amplify the site-specific region (Killebrew et al, 2004).


T-box 18. T-box 18 (TBX18). Forty-four transcripts with expression differences higher than 2-fold (T test, P< or =0.05) were detected between forelimb and hindlimb tissues including 38 new transcripts such as Rdh10, Frzb, Tbx18, and Hip that exhibit differential limb expression (Shou et al, 2005). T-box genes have been implicated in early cardiac lineage determination, chamber specification, valvuloseptal development, and diversification of the specialized conduction system in vertebrate embryos. These genes include Tbx1, Tbx2, Tbx3, Tbx5, Tbx18, and Tbx20, all of which exhibit complex temporal spatial regulation in developing cardiac structures (Plageman and Yutzey, 2005). It was demonstrated that maintenance of anterior-posterior-somite polarity is mediated by the T-box transcription factor Tbx18 (Bussen et al, 2004). Given the haploinsufficiency phenotypes reported for other T-box genes, we speculate that allelic imbalance (AI) may influence the function of Tbx18 during osteosarcomagenesis (Rosemann et al, 2004).


PLA2G3. PLA2G3 is also known as phospholipase A2 group III (GIII-SPLA2, sPLA(2)-III). Human group III secreted phospholipase A(2) (sPLA(2)-III) consists of a central group III sPLA(2) domain flanked by unique N- and C-terminal domains. It was found that the sPLA(2) domain alone was sufficient for its catalytic activity and for its prostaglandin E(2) (PGE(2))-generating functions in various cell types. Immunohistochemistry demonstrated that sPLA(2)-III was preferentially expressed in the microvascular endothelium in human tissues with inflammation, ischemic injury, and cancer. In support of this, sPLA(2)-III was induced in cultured microvascular endothelial cells after stimulation with proinflammatory cytokines. Expression of sPLA(2)-III was also associated with various tumor cells, and colorectal cancer cells transfected with sPLA(2)-III exhibited enhanced PGE(2) production and cell proliferation, which required sPLA(2)-III catalytic activity (Murakami et al, 2005).


OTTHUMG00000030140. OTTHUMG00000030140, CTA-299D3.6 (Vega gene ID) alias bA262A13.C22.5. Identification of the breakpoint between CTA-299D3 and RP5-925J7 probe, located in 22q13.32. Deletion extent could be estimated to be about 2.5 Mb in a patient with ring chromosome 22 (includes mental retardation with severe language impairment, hypotonia, and dysmorphic facial features) (Battini et al, 2004).


Solute carrier family 7 (cationic amino acid transporter, y+ system) member 4. Solute carrier family 7 (cationic amino acid transporter, y+ system) member 4 (SLC7A4, CAT4, CAT-4, HCAT3, MGC129976, MGC129977) exhibits significant sequence homology with the SLC7 subfamily of human cationic amino acid transporters (hCATs). Human glioblastoma cells stably overexpressing a fusion protein between SLC7A4 and the enhanced green fluorescent protein (EGFP) did not exhibit an increased transport activity for 1-arginine. The lack of transport activity was not due to a lack of SLC7A4 protein expression in the plasma membrane. The expression of SLC7A4 can be induced in NT2 teratocarcinoma cells by treatment with retinoic acid. However, also for this endogenously expressed SLC7A4, any transport activity for 1-arginine could not be detected. Therefore, SLC7A4 is either not an amino acid transporter or it needs additional (protein) factor(s) to be functional (Wolf et al, 2002). SLC7A4 was mapped to 22q11.2, the commonly deleted region of the velocardiofacial syndrome (VCFS, Shprintzen syndrome). In a patient affected by VCFS, deletion of SLC7A4 was demonstrated by chromosomal FISH. By Northern analysis, an abundant transcript was detected in brain, testis, and placenta (Sperandeo et al, 1998).


Sushi domain containing 2. Sushi domain containing 2 (SUSD2 alias BK65A6.2, FLJ22778). Neuronal marker proteins are widely used for characterization and identification of normal and tumor tissue of the central nervous system, but the most commonly used neuronal markers have inherent methodological problems. A proteomic approach was used comprising two-dimensional (2-D) gel electrophoresis and subsequent MALDI identification to identify possible new marker proteins in the human cortical neuronal cell line HCN-2. 14 proteins were found, among them BK65A6.2 (Novel Sushi Domain (Scr repeat)) (Peyrl et al, 2003).


Phosphatidylinositol (4,5) bisphosphate 5-phosphatase, A. Phosphatidylinositol (4,5) bisphosphate 5-phosphatase A (PIB5PA alias PIPP; INPP5; MGC129984 PIPP) hydrolyzes PtdIns(3,4,5)P3 forming PtdIns(3,4)P2, decreasing Ser473-Akt phosphorylation. PIPP is expressed in PC12 cells, localizing to the plasma membrane of undifferentiated cells and the neurite shaft and growth cone of NGF-differentiated neurites. Overexpression of wild-type, but not catalytically-inactive PIPP, in PC12 cells inhibited neurite elongation. Targeted depletion of PIPP using RNA interference (RNAi) resulted in enhanced neurite differentiation, associated with neurite hyper-elongation (Ooms et al 2005).


Signal peptide-CUB domian-EGF-related 1. Signal peptide-CUB domian-EGF-related 1 (SCUBE1) mRNA is found in several highly vascularized tissues such as liver, kidney, lung, spleen, and brain and is selectively expressed in vascular endothelial cells (EC) by in situ hybridization (Yang et al, 2002). The Scube1 (signal peptide-CUB domain-EGF-related 1) gene was isolated from a developing mouse urogenital ridge cDNA library and is expressed prominently in the developing gonad, nervous system, somites, surface ectoderm, and limb buds (Grimmond et al, 2000). SCUBE1 and SCUBE2 define an emerging family of human secreted proteins that are expressed in vascular endothelium and may play important roles in development, inflammation, and thrombosis (Yang et al, 2000).


RP3-355C18.2 (Vega gene ID). RP3-355C18.2 (Vega gene ID) alias dJ355C18.C22.2 is similar to AK023960 (according to Vega Report).


RP1-47A17.8 (Vega gene ID). RP1-47A17.8 (Vega gene ID) alias dJ47A17.C22.8 is similar to AL110226 cDNA DKFZp434H204 (According to Vega Report).


KB-1323B2.3. Embryonic marker KB-1323B2.3 alias Em:AP000557.C22.3 is similar to Em:AF012872 human phosphatidylinositol 4-kinase 230 (According to Vega Report).


Breast carcinoma amplified sequence 4. The breast carcinoma amplified sequence 4 (BCAS4 alias FLJ20495, BHLHB4) gene at 20q13.2 encodes a 211 amino acid cytoplasmic protein. In the MCF7 breast cancer cell line, the BCAS3 and BCAS4 genes were co-amplified, and cloning of a highly overexpressed 1.3-kb transcript revealed a rearrangement fusing the last two exons of BCAS3 with BCAS4. The BCAS4-BCAS3 fusion transcript was detected only in MCF7 cells, but the BCAS4 gene was also overexpressed in nine of 13 breast cancer cell lines (Barlund et al, 2004).


SAM50-like protein CGI-51. SAM50-like protein CGI-51 also known as SAM50_HUMAN (UniProt/Swiss-Prot ID) or sorting and assembly machinery component 50 homolog (S. Cerevisiae) (SAMM50 alias OMP85, SAM50, TOB55, TRG-3, CGI-51, YNL026W). Tob55 is an essential component of the TOB complex in that it constitutes the core element of the protein-conducting pore. The other two components of the TOB complex are Tob38, which builds a functional TOB core complex with Tob55, and Mas37, a peripheral member of the complex. Reduced insertion of the Tob55 precursor in the absence of Tom20 and Tom70 argues for initial recognition of the precursor of Tob55 by the import receptors. Next, it is transferred through the import channel formed by Tom40 (Habib et al, 2005).


TPX1_testis specific protein 1. TPX1_testis specific protein 1 (probe H4-1 p3-1) (CRISP2 alias TPX1, TSP1, GAPDL5, CRISP-2, MGC111136). TPX1 is a component of the sperm acrosome that remains associated with sperm after capacitation and acrosome reaction, and is relevant for sperm-oocyte interaction (Busso et al, 2004). There is significant difference in the expression levels of TPX-1 between normal (n=29) and motility impaired (n=24) semen samples, indicating that this gene is involved in sperm function (Wang et al, 2004). RT-PCR analysis of RNA isolated from acinar cells of lacrimal glands revealed that they expressed CRISP-1 and CRISP-2 (Haendler et al, 1999).


Nesprin-1. Nesprin-1 (Nuclear envelope spectrin repeat protein 1) is also known as synaptic nuclear envelope protein 1 (Syne-1), Myocyte nuclear envelope protein 1 (Myne-1) or Enaptin (SYNE1 alias SYNE-1B, KIAA0796, 8B, nesprin-1, enaptin, MYNE1, CPG2). Transgenic mice overexpressing the conserved C-terminal Klarsicht/ANC-1/Syne homology domain of Syne-1 were generated. The transgene acted in a dominant interfering fashion, displacing endogenous Syne-1 from the nuclear envelope. Muscle nuclei failed to aggregate at the NMJ in transgenic mice, demonstrating that localization and positioning of synaptic nuclei require Syne proteins (Grady et al, 2005). Integral membrane protein nesprin-1alpha serves as a receptor for mAKAP on the nuclear envelope in cardiac myocytes (Pare et al, 2005). Syne-1 and KIF3B function together in cytokinesis by facilitating the accumulation of membrane vesicles at the spindle midbody (Fan and Beck, 2004). Syne-1 gene is expressed in a variety of forms that are multifunctional and are capable of functioning at both the Golgi and the nuclear envelope, perhaps linking the two organelles during muscle differentiation (Gough et al, 2003). Nesprin-1 is developmentally regulated in both smooth and skeletal muscle and is re-localized from the nuclear envelope to the nucleus and cytoplasm during C2C12 myoblast differentiation (Zhang et al, 2001).


FLOT1. FLOT1 is also known as flotillin or 1 ENSG00000137312. Caveolae are small domains on the inner cell membrane involved in vesicular trafficking and signal transduction. FLOT1 encodes a caveolae-associated, integral membrane protein. The function of flotillin 1 has not been determined (Entrez database).


C6 orf25. C6orf25 is also known as chromosome 6 open reading frame 25; ENSG00000096148; C6orf25; G6b; NG31. This gene is a member of the immunoglobulin (Ig) superfamily and is located in the major histocompatibility complex (MHC) class III region. The protein encoded by this gene is a glycosylated, plasma membrane-bound cell surface receptor, but soluble isoforms encoded by some transcript variants have been found in the endoplasmic reticulum and Golgi before being secreted. Seven transcript variants encoding different isoforms have been described for this gene (Entrez database).


VARS. VARS is also known as valyl-tRNA synthetase; ENSG00000096171; G7A or VARS2.


Aminoacyl-tRNA synthetases catalyze the aminoacylation of tRNA by their cognate amino acid. Because of their central role in linking amino acids with nucleotide triplets contained in tRNAs, aminoacyl-tRNA synthetases are thought to be among the first proteins that appeared in evolution. The protein encoded by this gene belongs to class-I aminoacyl-tRNA synthetase family and is located in the class III region of the major histocompatibility complex (Entrez database).


Major histocompatibility complex, class II, DP beta 1. Major histocompatibility complex, class II, DP beta 1 is also known as OTTHUMG00000031076; HLA-DPB1; HLA-DPB1; DPB1; HLA-DP1 B; MHC DPB1. HLA-DPB belongs to the HLA class II beta chain paralogues. This class II molecule is a heterodimer consisting of an alpha (DPA) and a beta chain (DPB), both anchored in the membrane. It plays a central role in the immune system by presenting peptides derived from extracellular proteins. Class II molecules are expressed in antigen presenting cells (APC: B lymphocytes, dendritic cells, macrophages). The beta chain is approximately 26-28 kDa and its gene contains 6 exons. Exon one encodes the leader peptide, exons 2 and 3 encode the two extracellular domains, exon 4 encodes the transmembrane domain and exon 5 encodes the cytoplasmic tail. Within the DP molecule both the alpha chain and the beta chain contain the polymorphisms specifying the peptide binding specificities, resulting in up to 4 different molecules (Entrez database).


Major histocompatibility complex, class II, DR beta 5. Major histocompatibility complex, class II, DR beta 5 is also known as OTTHUMG00000031027; HLA-DRB5. HLA-DRB5 belongs to the HLA class II beta chain paralogues. This class II molecule is a heterodimer consisting of an alpha (DRA) and a beta (DRB) chain, both anchored in the membrane. It plays a central role in the immune system by presenting peptides derived from extracellular proteins. Class II molecules are expressed in antigen presenting cells (APC: B lymphocytes, dendritic cells, macrophages). The beta chain is approximately 26-28 kDa and its gene contains 6 exons. Exon one encodes the leader peptide, exons 2 and 3 encode the two extracellular domains, exon 4 encodes the transmembrane domain and exon 5 encodes the cytoplasmic tail. Within the DR molecule the beta chain contains all the polymorphisms specifying the peptide binding specificities. Typing for these polymorphisms is routinely done for bone marrow and kidney transplantation. DRB1 is expressed at a level five times higher than its paralogues DRB3, DRB4 and DRB5. The presence of DRB5 is linked with allelic variants of DRB1, otherwise it is omitted. There are 4 related pseudogenes: DRB2, DRB6, DRB7, DRB8 and DRB9 (Entrez database).


COL11A2. COL11A2 is also known as collagen, type XI, alpha 2; OTTHUMG00000031036; HKE5; PARP; STL3; DFNA13; DFNB53. This gene encodes one of the two alpha chains of type XI collagen, a minor fibrillar collagen. It is located on chromosome 6 very close to but separate from the gene for retinoid X receptor beta. Type XI collagen is a heterotrimer but the third alpha chain is a post-translationally modified alpha 1 type II chain. Proteolytic processing of this type XI chain produces PARP, a proline/arginine-rich protein that is an amino terminal domain. Mutations in this gene are associated with type III Stickler syndrome, otospondylomegaepiphyseal dysplasia (OSMED syndrome), Weissenbacher-Zweymuller syndrome, and autosomal dominant nonsyndromic sensorineural 13 deafness. Three transcript variants encoding different isoforms have been identified for this gene (Entrez database).


PRAME. PRAME is also known as melanoma antigen preferentially expressed in tumors; preferentially expressed antigen of melanoma or OPA-interacting protein 4 (OIP4 alias ENSG0000185686; MAPE). This gene encodes an antigen that is predominantly expressed in human melanomas and that is recognized by cytolytic T lymphocytes. It is not expressed in normal tissues, except testis. This expression pattern is similar to that of other CT antigens, such as MAGE, BAGE and GAGE. However, unlike these other CT antigens, this gene is also expressed in acute leukemias. Five alternatively spliced transcript variants encoding the same protein have been observed for this gene (Entrez database).


FBLN1. FBLN1 is also known as fibulin 1 or ENSG00000077942. Fibulin 1 is a secreted glycoprotein that becomes incorporated into a fibrillar extracellular matrix. Calcium-binding is apparently required to mediate its binding to laminin and nidogen. It mediates platelet adhesion via binding fibrinogen. Four splice variants which differ in the 3′ end have been identified. Each variant encodes a different isoform, but no functional distinctions have been identified among the four variants (Entrez database).


CYP2D6. CYP2D6 is also known as cytochrome P450, family 2, subfamily D, polypeptide 6 or ENSG00000100197 (alias CPD6; CYP2D; CYP2D@; CYP2DL1; P450C2D; P450-DB1; MGC120389; MGC120390). This gene encodes a member of the cytochrome P450 superfamily of enzymes. The cytochrome P450 proteins are monooxygenases which catalyze many reactions involved in drug metabolism and synthesis of cholesterol, steroids and other lipids. This protein localizes to the endoplasmic reticulum and is known to metabolize as many as 20% of commonly prescribed drugs. Its substrates include debrisoquine, an adrenergic-blocking drug; sparteine and propafenone, both anti-arrythmic drugs; and amitryptiline, an anti-depressant. The gene is highly polymorphic in the population; certain alleles result in the poor metabolizer phenotype, characterized by a decreased ability to metabolize the enzyme's substrates. The gene is located near two cytochrome P450 pseudogenes on chromosome 22q13.1. Alternatively spliced transcript variants encoding different isoforms have been found for this gene (Entrez database).


AC006548.8 (Vega gene ID). AC006548.8 (Vega gene ID) is also known as Em:AC006548.C22.8. This gene is similar to Em:AB018413 human Gab2 (According to Vega Report).



Homo sapiens cat eye syndrome critical region 3 (CECR3) gene. Homo sapiens cat eye syndrome critical region 3 (CECR3) gene (According to Vega Report) is also known as topoisomerase (DNA) III beta, OTTHUMG00000030764 or TOP3B. This gene encodes a DNA topoisomerase, an enzyme that controls and alters the topologic states of DNA during transcription. This enzyme catalyzes the transient breaking and rejoining of a single strand of DNA which allows the strands to pass through one another, thus relaxing the supercoils and altering the topology of DNA. The enzyme interacts with DNA helicase SGS1 and plays a role in DNA recombination, cellular aging and maintenance of genome stability. Alternative splicing of the C-terminus of this gene results in three transcript variants which have distinct tissue specificity; however, not all variants have been fully described (Entrez database).


KB-1269D1.3 (Vega gene ID). KB-1269D1.3 (Vega gene ID) is also known as OTTHUMG00000030694 or Em:AP000344.C22.3. It is similar to Tr:Q13312 human TXBP181 (According to Vega Report).


GPR24. GPR24 is also known as G protein-coupled receptor 24; ENSG00000128285; SLC1; MCHR1; MGC32129. The protein encoded by this gene, a member of the G protein-coupled receptor family 1, is an integral plasma membrane protein which binds melanin-concentrating hormone. The encoded protein can inhibit cAMP accumulation and stimulate intracellular calcium flux, and is probably involved in the neuronal regulation of food consumption. Although structurally similar to somatostatin receptors, this protein does not seem to bind somatostatin (Entrez database).


GAL3ST1 GAL3ST1 is also known as galactose-3-O-sulfotransferase 1; ENSG00000128242; CST. Sulfonation, an important step in the metabolism of many drugs, xenobiotics, hormones, and neurotransmitters, is catalyzed by sulfotransferases. The product of this gene is galactosylceramide sulfotransferase which catalyzes the conversion between 3′-phosphoadenylylsulfate+a galactosylceramide to adenosine 3′,5′-bisphosphate+galactosylceramide sulfate. Activity of this sulfotransferase is enhanced in renal cell carcinoma (Entrez database).


GSTT3-3 similar to Glutathione S-transferases (according to Vega Report). GSTT3-3 similar to Glutathione S-transferases (according to Vega Report) is also known as RP4-539M6.7 (Vega gene ID); OTTHUMG00000030918 or Em:AC004832.C22.7. It is similar to TR:Q9Y2Z7 Homo sapiens CGI-08 PROTEIN (according to Vega Report).


GALR3GALR3 is also known as galanin receptor 3 or ENSG00000128310. The neuropeptide galanin modulates a variety of physiologic processes including cognition/memory, sensory/pain processing, hormone secretion, and feeding behavior. The human galanin receptors are G protein-coupled receptors that functionally couple to their intracellular effector through distinct signaling pathways. GALR3 is found in many tissues and may be expressed as 1.4-, 2.4-, and 5-kb transcripts (Entrez database).


IL2RB. IL2RB is also known as interleukin 2 receptor, beta; ENSG00000100385 or P70-75. The interleukin 2 receptor, which is involved in T cell-mediated immune responses, is present in 3 forms with respect to ability to bind interleukin 2. The low affinity form is a monomer of the alpha subunit and is not involved in signal transduction. The intermediate affinity form consists of an alpha/beta subunit heterodimer, while the high affinity form consists of an alpha/beta/gamma subunit heterotrimer. Both the intermediate and high affinity forms of the receptor are involved in receptor-mediated endocytosis and transduction of mitogenic signals from interleukin 2. The protein encoded by this gene represents the beta subunit and is a type I membrane protein (Entrez database).


DGCR2. DGCR2 is also known as DiGeorge syndrome critical region gene 2; ENSG00000070413; IDD; LAN; DGS-C; SEZ-12; KIAA0163; DKFZp68611730. Deletions of the 22q11.2 have been associated with a wide range of developmental defects (notably DiGeorge syndrome, velocardiofacial syndrome, conotruncal anomaly face syndrome and isolated conotruncal cardiac defects) classified under the acronym CATCH 22. The DGCR2 gene encodes a novel putative adhesion receptor protein, which could play a role in neural crest cells migration, a process which has been proposed to be altered in DiGeorge syndrome (Entrez database).


TCN2. TCN2 is also known as transcobalamin II; macrocytic anemia; ENSG00000185339 (alias TC2; D22S676; D22S750). This gene encodes a member of the vitamin B12-binding protein family. This family of proteins, alternatively referred to as R binders, is expressed in various tissues and secretions. This plasma protein binds cobalamin and mediates the transport of cobalamin into cells. This protein and other mammalian cobalamin-binding proteins, such as transcobalamin I and gastric intrisic factor, may have evolved by duplication of a common ancestral gene (Entrez database).


IGLL1. IGLL1 is also known as immunoglobulin lambda-like polypeptide 1 or ENSG00000128322 (alias IGO; 14.1; IGL1; IGL5; IGLL; IGVPB; CD179b; VPREB2; IGLJ14.1). The preB cell receptor is found on the surface of proB and preB cells, where it is involved in transduction of signals for cellular proliferation, differentiation from the proB cell to the preB cell stage, allelic exclusion at the Ig heavy chain gene locus, and promotion of Ig light chain gene rearrangements. The preB cell receptor is composed of a membrane-bound Ig mu heavy chain in association with a heterodimeric surrogate light chain. This gene encodes one of the surrogate light chain subunits and is a member of the immunoglobulin gene superfamily. This gene does not undergo rearrangement. Mutations in this gene can result in B cell deficiency and agammaglobulinemia, an autosomal recessive disease in which few or no gamma globulins or antibodies are made. Two transcript variants encoding different isoforms have been found for this gene (Entrez database).


APOBEC3B. APOBEC3B is also known as apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3B; ENSG00000179750 (alias ARP4; ARCD3; PHRBNL; APOBEC1L; FLJ21201; DJ742C19.2). This gene is a member of the cytidine deaminase gene family. It is one of seven related genes or pseudogenes found in a cluster, thought to result from gene duplication, on chromosome 22. Members of the cluster encode proteins that are structurally and functionally related to the C to U RNA-editing cytidine deaminase APOBEC1. It is thought that the proteins may be RNA editing enzymes and have roles in growth or cell cycle control (Entrez database).


CRYBB1. CRYBB1 is also known as crystallin, beta B1 or ENSG00000100122. Crystallins are separated into two classes: taxon-specific, or enzyme, and ubiquitous. The latter class constitutes the major proteins of vertebrate eye lens and maintains the transparency and refractive index of the lens. Since lens central fiber cells lose their nuclei during development, these crystallins are made and then retained throughout life, making them extremely stable proteins. Mammalian lens crystallins are divided into alpha, beta, and gamma families; beta and gamma crystallins are also considered as a superfamily. Alpha and beta families are further divided into acidic and basic groups. Seven protein regions exist in crystallins: four homologous motifs, a connecting peptide, and N- and C-terminal extensions. Beta-crystallins, the most heterogeneous, differ by the presence of the C-terminal extension (present in the basic group, none in the acidic group). Beta-crystallins form aggregates of different sizes and are able to self-associate to form dimers or to form heterodimers with other beta-crystallins. This gene, a beta basic group member, undergoes extensive cleavage at its N-terminal extension during lens maturation. It is also a member of a gene cluster with beta-A4, beta-B2, and beta-B3 (Entrez database).


CRYBA4. CRYBA4 is also known as crystallin, beta A4 or ENSG00000196431. Crystallins are separated into two classes: taxon-specific, or enzyme, and ubiquitous. The latter class constitutes the major proteins of vertebrate eye lens and maintains the transparency and refractive index of the lens. Since lens central fiber cells lose their nuclei during development, these crystallins are made and then retained throughout life, making them extremely stable proteins. Mammalian lens crystallins are divided into alpha, beta, and gamma families; beta and gamma crystallins are also considered as a superfamily. Alpha and beta families are further divided into acidic and basic groups. Seven protein regions exist in crystallins: four homologous motifs, a connecting peptide, and N- and C-terminal extensions. Beta-crystallins, the most heterogeneous, differ by the presence of the C-terminal extension (present in the basic group, none in the acidic group). Beta-crystallins form aggregates of different sizes and are able to self-associate to form dimers or to form heterodimers with other beta-crystallins. This gene, a beta acidic group member, is part of a gene cluster with beta-B1, beta-B2, and beta-B3 (Entrez database).


APOL4. APOL4 is also known as apolipoprotein L 4 or ENSG00000100336 (alias APOLIV; APOL-IV). The protein encoded by this gene is a member of the apolipoprotein L family and may play a role in lipid exchange and transport throughout the body, as well as in reverse cholesterol transport from peripheral cells to the liver. Two transcript variants encoding two different isoforms have been found for this gene. Only one of the isoforms appears to be a secreted protein (Entrez database).


SOX10. SOX10 is also known as SRY (sex determining region Y)-box 10 or ENSG00000100146 (alias DOM; WS4; MGC15649). This gene encodes a member of the SOX (SRY-related HMG-box) family of transcription factors involved in the regulation of embryonic development and in the determination of the cell fate. The encoded protein may act as a transcriptional activator after forming a protein complex with other proteins. This protein acts as a nucleocytoplasmic shuttle protein and is important for neural crest and peripheral nervous system development. Mutations in this gene are associated with Waardenburg-Shah and Waardenburg-Hirschsprung disease (Entrez database).


MGAT3. MGAT3 is also known as mannosyl (beta-1,4-)-glycoprotein beta-1,4-N-acetylglucosaminyltransferase or ENSG00000128268 (alias GNT3; GNT-III). There are believed to be over 100 different glycosyltransferases involved in the synthesis of protein-bound and lipid-bound oligosaccharides. N-acetylglucosaminyltransferase III transfers a GlcNAc residue to the beta-linked mannose of the trimannosyl core of N-linked oligosaccharides and produces a bisecting GlcNAc. Expression of this gene may be controlled by a multiple-promoter system (Entrez database).


RABL4. RABL4 is also known as RAB member of RAS oncogene family-like 4 or ENSG00000100360 (alias RAYL). This gene encodes a putative GTP-binding protein similar to RAY/RAB1C. The protein is ras-related, but the function is unknown (Entrez database).


SULT4A1. SULT4A1 is also known as sulfotransferase family 4A, member 1; or ENSG00000130540 (alias NST; BRSTL1; SULTX3; BR-STL-1; MGC40032; DJ388M5.3; hBR-STL-1). Sulfotransferase enzymes catalyze the sulfate conjugation of many hormones, neurotransmitters, drugs, and xenobiotic compounds. These cytosolic enzymes are different in their tissue distributions and substrate specificities. The gene structure (number and length of exons) is similar among family members. This gene encodes a protein that is selectively expressed in brain tissue (Entrez database).


RPL3. RPL3 is also known as ribosomal protein L3 or ENSG00000100316 alias (TARBP-B; MGC104284). Ribosomes, the complexes that catalyze protein synthesis, consist of a small 40S subunit and a large 60S subunit. Together these subunits are composed of 4 RNA species and approximately 80 structurally distinct proteins. This gene encodes a ribosomal protein that is a component of the 60S subunit. The protein belongs to the L3P family of ribosomal proteins and it is located in the cytoplasm. The protein can bind to the HIV-1 TAR mRNA, and it has been suggested that the protein contributes to tat-mediated transactivation. This gene is co-transcribed with several small nucleolar RNA genes, which are located in several introns of this gene. Alternate transcriptional splice variants, encoding different isoforms, have been characterized. As is typical for genes encoding ribosomal proteins, there are multiple processed pseudogenes of this gene dispersed through the genome (Entrez database).


APOL2. APOL2 is also known as apolipoprotein L, 2 or ENSG00000128335 (alias APOL-II). This gene is a member of the apolipoprotein L gene family. The encoded protein is found in the cytoplasm, where it may affect the movement of lipids or allow the binding of lipids to organelles. Two transcript variants encoding the same protein have been found for this gene (Entrez database).


RAC2. RAC2 is also known as ras-related C3 botulinum toxin substrate 2 (rho family, small GTP binding protein Rac2) or ENSG00000128340 (alias Gx; EN-7; HSPC022). The protein encoded by this gene is a GTPase which belongs to the RAS superfamily of small GTP-binding proteins. Members of this superfamily appear to regulate a diverse array of cellular events, including the control of cell growth, cytoskeletal reorganization, and the activation of protein kinases (Entrez database).


GABRR2. GABRR2 is also known as gamma-aminobutyric acid (GABA) receptor, rho 2 or ENSG00000111886. GABA is the major inhibitory neurotransmitter in the mammalian brain where it acts at GABA receptors, which are ligand-gated chloride channels. GABRR2 is a member of the rho subunit family (Entrez database).


MOG. MOG is also known as myelin oligodendrocyte glycoprotein or ENSG00000137345 (alias MGC26137). The product of this gene is a membrane protein expressed on the oligodendrocyte cell surface and the outermost surface of myelin sheaths. Due to this localization, it is a primary target antigen involved in immune-mediated demyelination. This protein may be involved in completion and maintenance of the myelin sheath and in cell-cell communication. Alternatively spliced transcript variants encoding different isoforms have been identified (Entrez database).


ME1. ME1 is also known as malic enzyme 1, NADP(+)-dependent, cytosolic or ENSG00000065833 (alias MES; HUMNDME). This gene encodes a cytosolic, NADP-dependent enzyme that generates NADPH for fatty acid biosynthesis. The activity of this enzyme, the reversible oxidative decarboxylation of malate, links the glycolytic and citric acid cycles. The regulation of expression for this gene is complex. Increased expression can result from elevated levels of thyroid hormones or by higher proportions of carbohydrates in the diet (Entrez database).


IL20RA. IL20RA is also known as interleukin 20 receptor, alpha or ENSG00000016402 (alias IL-20R1; ZCYTOR7). The protein encoded by this gene is a receptor for interleukin 20 (IL20), a cytokine that may be involved in epidermal function. The receptor of IL20 is a heterodimeric receptor complex consisting of this protein and interleukin 20 receptor beta (IL20B). This gene and IL20B are highly expressed in skin. The expression of both genes is found to be upregulated in Psoriasis (Entrez database).


ZHX3. ZHX3 is also known as zinc fingers and homeoboxes 3 or OTTHUMG00000032481 (alias TIX1; KIAA0395). This gene encodes a member of the zinc fingers and homeoboxes (ZHX) gene family. The encoded protein contains two C2H2-type zinc fingers and five homeodomains and forms a dimer with itself or with zinc fingers and homeoboxes family member 1. In the nucleus, the dimerized protein interacts with the A subunit of the ubiquitous transcription factor nuclear factor-Y and may function as a transcriptional repressor (Entrez database).


CHD6. CHD6 is also known as chromodomain helicase DNA binding protein 6 or ENSG00000124177 (alias CHD5; RIGB; KIAA1335). Chromosomal DNA of eukaryotic cells is compacted by nuclear proteins to form chromatin, an organized nucleoprotein structure that can inhibit gene expression. Several multisubunit protein complexes exist to remodel the chromatin to allow patterns of cell type-specific gene expression. The protein encoded by this gene is thought to be a core member of one or more of these complexes. The encoded protein, which is a member of the SNF2/RAD54 helicase family, contains two chromodomains, a helicase domain, and an ATPase domain (Entrez database).


PTPRG. PTPRG is also known as protein tyrosine phosphatase, receptor type, 6 or ENSG00000144724 (alias PTPG; HPTPG; RPTPG; R-PTP-GAMMA). The protein encoded by this gene is a member of the protein tyrosine phosphatase (PTP) family. PTPs are known to be signaling molecules that regulate a variety of cellular processes including cell growth, differentiation, mitotic cycle, and oncogenic transformation. This PTP possesses an extracellular region, a single transmembrane region, and two tandem intracytoplasmic catalytic domains, and thus represents a receptor-type PTP. The extracellular region of this PTP contains a carbonic anhydrase-like (CAH) domain, which is also found in the extracellular region of PTPRBETA/ZETA. This gene is located in a chromosomal region that is frequently deleted in renal cell carcinoma and lung carcinoma, thus is thought to be a candidate tumor suppressor gene (Entrez database).


PTPNS1. PTPNS1 is also known as protein tyrosine phosphatase, non-receptor type substrate 1 or ENSG00000198053 (alias BIT; MFR; P84; SIRP; MYD-1; SHPS1; SHPS-1; SIRPalpha; SIRPalpha2; SIRP-ALPHA-1). The protein encoded by this gene is a member of the signal-regulatory-protein (SIRP) family, and also belongs to the immunoglobulin superfamily. SIRP family members are receptor-type transmembrane glycoproteins known to be involved in the negative regulation of receptor tyrosine kinase-coupled signaling processes. This protein can be phosphorylated by tyrosine kinases. The phospho-tyrosine residues of this PTP have been shown to recruit SH2 domain containing tyrosine phosphatases (PTP), and serve as substrates of PTPs. This protein was found to participate in signal transduction mediated by various growth factor receptors. CD47 has been demonstrated to be a ligand for this receptor protein. This gene and its product share very high similarity with several other members of the SIRP family. These related genes are located in close proximity to each other on chromosome 20p13 (Entrez database).


PCSK2. PCSK2 is also known as proprotein convertase subtilisin/kexin type 2 or ENSG00000125851 (alias PC2; NEC2; SPC2). The protein encoded by this gene belongs to the subtilisin-like proprotein convertase family. The members of this family are proprotein convertases that process latent precursor proteins into their biologically active products. This encoded protein is a proinsulin-processing enzyme that plays a key role in regulating insulin biosynthesis. It is also known to cleave proopiomelanocortin, proenkephalin, prodynorphin and proluteinizing-hormone-releasing hormone. The use of alternate polyadenylation sites has been found for this gene (Entrez database).


PLAGL2. PLAGL2 is also known as pleiomorphic adenoma gene-like 2 or ENSG00000126003. It is a zinc-finger protein that recognizes DNA and/or RNA (Entrez database).


GGTL3. GGTL3 is also known as gamma-glutamyltransferase-like 3 or ENSG00000131067 (alias GGTL5; D20S101; dJ18C9.2). Gamma-glutamyltransferase is a membrane-associated protein involved in both the metabolism of glutathione and in the transpeptidation of amino acids. Changes in the activity of gamma-glutamyltransferase may signal preneoplastic or toxic conditions in the liver or kidney. The protein encoded by this gene is similar in sequence to gamma-glutamyltransferase, but its function is unknown. Multiple transcript variants encoding several different isoforms have been found for this gene (Entrez database).


EPB41L1. EPB41L1 is also known as erythrocyte membrane protein band 4.1-like 1 or ENSG00000088367 (alias KIAA0338; MGC11072). Erythrocyte membrane protein band 4.1 (EPB41) is a multifunctional protein that mediates interactions between the erythrocyte cytoskeleton and the overlying plasma membrane. The protein encoded by this gene is a neuronally-enriched protein that is structurally similar to EPB41. The encoded protein binds and stabilizes D2 and D3 dopamine receptors at the neuronal plasma membrane. Multiple transcript variants encoding different isoforms have been found for this gene, but the full-length nature of only two of them has been determined (Entrez database).


SDC4. SDC4 is also known as syndecan 4 (amphighlycan, ryudocan) or ENSG00000124145 (alias SYND4; MGC22217). The protein encoded by this gene is a transmembrane (type 1) heparan sulfate proteoglycan that functions as a receptor in intracellular signaling. The encoded protein is found as a homodimer and is a member of the syndecan proteoglycan family. This gene is found on chromosome 20, while a pseudogene has been found on chromosome 22 (Entrez database).


EYA2. EYA2 is also known as eyes absent homolog 2 (Drosophila) or ENSG00000064655 (alias EAB1; MGC10614). This gene encodes a member of the eyes absent (EYA) family of proteins. The encoded protein may be post-translationally modified and may play a role in eye development. A similar protein in mice can act as a transcriptional activator. Five transcript variants encoding three distinct isoforms have been identified for this gene (Entrez database).


USP18. USP18 is also known as ubiquitin specific peptidase 18 or OTTHUMG00000030949 (alias ISG43; UBP43). USP18, a member of the deubiquitinating protease family of enzymes, removes ubiquitin adducts from a broad range of protein substrates (supplied by OMIM).


BCR. BCR is also known as breakpoint cluster region or ENSG00000186716 (alias ALL; CML; PHL; BCR1; D22S11; D22S662). A reciprocal translocation between chromosomes 22 and 9 produces the Philadelphia chromosome, which is often found in patients with chronic myelogenous leukemia. The chromosome 22 breakpoint for this translocation is located within the BCR gene. The translocation produces a fusion protein which is encoded by sequence from both BCR and ABL, the gene at the chromosome 9 breakpoint. Although the BCR-ABL fusion protein has been extensively studied, the function of the normal BCR gene product is not clear. The protein has serine/threonine kinase activity and is a GTPase-activating protein for p21rac. Two transcript variants encoding different isoforms have been found for this gene (Entrez database).


MAPK8IP2. MAPK8IP2 is also known as mitogen-activated protein kinase 8 interacting protein 2 or ENSG00000008735 (alias IB2; JIP2; PRKM8IPL). The protein encoded by this gene is closely related to MAPK8IP1/IB1/JIP-1, a scaffold protein that is involved in the c-Jun amino-terminal kinase signaling pathway. This protein is expressed in brain and pancreatic cells. It has been shown to interact with, and regulate the activity of MAPK8/JNK1, and MAP2K7/MKK7 kinases. This protein thus is thought to function as a regulator of signal transduction by protein kinase cascade in brain and pancreatic beta-cells. Three alternatively spliced transcript variants of this gene encoding distinct isoforms have been reported (Entrez database).


MN1. MN1 is also known as meningioma (disrupted in balanced translocation) 1 or ENSG00000169184 (alias MGCR; MGCR1; MGCR1-PEN; dJ353E16.2). Meningioma 1 (MN1) contains two sets of CAG repeats. It is disrupted by a balanced translocation (4;22) in a meningioma, and its inactivation may contribute to meningioma 32 pathogenesis (Entrez database).


RTDR1. RTDR1 is also known as rhabdoid tumor deletion region gene 1 or ENSG00000100218 (alias MGC16968). This gene encodes a protein with no known function but with slight similarity to a yeast vacuolar protein. The gene is located in a region deleted in pediatric rhabdoid tumors of the brain, kidney and soft tissues, but mutations in this gene have not been associated with the disease (Entrez database).


Solute carrier family 35 member E4. Solute carrier family 35 member E4 is also known as SLC35E4.


Glycoprotein Ib (platelet) beta polypeptide. Glycoprotein Ib (platelet) beta polypeptide CTA-243E7.3. CTA-243E7.3 is also known as bK243E7.C22.3.


CT026_HUMAN. CT026_HUMAN is also known as C20orf26, dJ1178H5.4; DKFZP434K156.


HIST1H3A. HIST1H3A (histone; H3/A alias H3FA). Histones are basic nuclear proteins that are responsible for the nucleosome structure of the chromosomal fiber in eukaryotes. This structure consists of approximately 146 bp of DNA wrapped around a nucleosome, an octamer composed of pairs of each of the four core histones (H2A, H2B, H3, and H4). The chromatin fiber is further compacted through the interaction of a linker histone, H1, with the DNA between the nucleosomes to form higher order chromatin structures. This gene is intronless and encodes a member of the histone H3 family. Transcripts from this gene lack polyA tails; instead, they contain a palindromic termination element. This gene is found in the large histone gene cluster on chromosome 6p22-p21.3 (Entrez).


Sorting nexin 5. Sorting nexin 5 (SNX5 alias FLJ10931) is a member of the sorting nexin family. Members of this family contain a phox (PX) domain, which is a phosphoinositide binding domain, and are involved in intracellular trafficking. This protein binds to fanconi anemia complementation group A protein, but its function is unknown. This gene results in two transcript variants encoding the same protein (Entrez).


Thioredoxin reductase 2 mitochondrial precursor. Thioredoxin reductase 2 mitochondrial precursor (EC 1.8.1.9) (TR3,TR-beta) is also known as selenoprotein Z (SelZ) alias TXNRD2, TR, TR3, SELZ, TRXR2, TR-BETA. Thioredoxin reductase (TR) is a dimeric NADPH-dependent FAD containing enzyme that catalyzes the reduction of the active site disulfide of thioredoxin and other substrates. TR is a member of a family of pyridine nucleotide-disulfide oxidoreductases and is a key enzyme in the regulation of the intracellular redox environment. Three thioredoxin reductase genes have been found that encode selenocysteine containing proteins. Alternative splicing of this gene results in three distinct transcripts encoding different isoforms, one of which has been shown to be located in the mitochondria. This TR gene partially overlaps the COMT gene on chromosome 22.


RP11-554D15.1. RP11-554D15.1 is also known as bA554D15.1.


BAI1-associated protein 2-like 2. BAI1-associated protein 2-like 2 is also known as BAIAP2L2 or FLJ22582.


Smoothelin. Smoothelin (SMTN) encodes a structural protein that is found exclusively in contractile smooth muscle cells. It associates with stress fibers and constitutes part of the cytoskeleton. This gene is localized to chromosome 22q12.3, distal to the TUPLE1 locus and outside the DiGeorge syndrome deletion. Alternative splicing of this gene results in three transcript variants (Entrez database).


Cadherin-4 precursor. Cadherin-4 precursor (CDH4, CAD4, RCAD, FLJ22202, FLJ40547, MGC126700) is also known as cadherin 4 type 1, Retinal-cadherin or R-cadherin (R-CAD). This gene is a classical cadherin from the cadherin superfamily. The encoded protein is a calcium-dependent cell-cell adhesion glycoprotein comprised of five extracellular cadherin repeats, a transmembrane region and a highly conserved cytoplasmic tail. Based on studies in chicken and mouse, this cadherin is thought to play an important role during brain segmentation and neuronal outgrowth. In addition, a role in kidney and muscle development is indicated. Of particular interest are studies showing stable cis-heterodimers of cadherins 2 and 4 in cotransfected cell lines. Previously thought to interact in an exclusively homophilic manner, this is the first evidence of cadherin heterodimerization (Entrez database).


Forkhead box protein O3A. Forkhead box protein O3A (FOXO3A, AF6q21, FKHRL1, FKHRL1P2, MGC12739, MGC31925, DKFZp781A0677) belongs to the forkhead family of transcription factors which are characterized by a distinct forkhead domain. This gene likely functions as a trigger for apoptosis through expression of genes necessary for cell death. Translocation of this gene with the MLL gene is associated with secondary acute leukemia. Alternatively spliced transcript variants encoding the same protein have been observed (Entrez database).


N-acetyllactosaminide beta-1,6-N-acetylglucosaminyl-transferase. The enzyme n-acetyllactosaminide beta-1,6-N-acetylglucosaminyl-transferase (EC 2.4.1.150; GCNT2, II GCNT5, II IGNT, ULG3, AIGnT, BIGnT, CIGnT, GCNT5, NAGCT1, bA421M1.1, bA360019.2) is responsible for the formation of the blood group I antigen. The i and I antigens are determined by linear and branched poly-N-acetyllactosaminoglycans, respectively. During embryonic development in human erythrocytes, the fetal i antigen is replaced by the adult I antigen as the result of the appearance of a beta-1,6-N-acetylglucosaminyltransferase, the I-branching enzyme. This gene encodes the I-branching enzyme that converts the linear form into the branched form. Defects in this gene have been associated with adult i blood group phenotype. Several transcript variants encoding different isoforms have been described (Entrez database).


Gamma-aminobutyric-acid receptor rho-1 subunit precursor. Gamma-aminobutyric-acid receptor rho-1 subunit precursor (GABRR1) is also known as GABA(A) receptor. GABA is the major inhibitory neurotransmitter in the mammalian brain where it acts at GABA receptors, which are ligand-gated chloride channels. GABRR1 is a member of the rho subunit family (Entrez database).


OTTHUMG00000030167. OTTHUMG00000030167 is also known as CTA-243E7.3 (Vega gene ID) or bK243E7.C22.3.


OTTHUMG00000030870. OTTHUMG00000030870 is also known as CTA-503F6.1 (Vega gene ID) or bK503F6.C22.1.


OTTHUMG00000030991. OTTHUMG00000030991 is also known as LL22NC03-75B3.6 (Vega gene ID), dJ671O14.C22.6 or KIAA1644.


Pantetheinase precursor. Pantetheinase precursor (EC 3.5.1.-; VNN1 alias Tiff66; MGC116930; MGC116931; MGC116932; MGC116933) is a member of the Vanin family of proteins which share extensive sequence similarity with each other, and also with biotinidase. The family includes secreted and membrane-associated proteins, a few of which have been reported to participate in hematopoietic cell trafficking. No biotinidase activity has been demonstrated for any of the vanin proteins, however, they possess pantetheinase activity, which may play a role in oxidative-stress response. This protein, like its mouse homolog, is likely a GPI-anchored cell surface molecule. The mouse protein is expressed by the perivascular thymic stromal cells and regulates migration of T-cell progenitors to the thymus. This gene lies in close proximity to, and in same transcriptional orientation as two other vanin genes on chromosome 6q23-q24 (Entrez database).


Mitochondrial glutamate carrier 2. Mitochondrial glutamate carrier 2 is also known as Glutamate/H(+) symporter 2 (GC2) or Solute carrier family 25 member 18 (SLC25A18).


OTTHUMG00000015693. OTTHUMG00000015693 is also known as RP11-12A2.3 (Vega_gene ID) or bA12A2.3.


RP5-899B16.1. RP5-899B16.1 is also known as dJ899B16.1.


Nuclear receptor coactivator 7. Nuclear receptor coactivator 7 (NCOA7 alias ESNA1; ERAP140; MGC88425; Nbla00052; Nbla10993; dJ187J11.3).


Protein phosphatase 1 regulatory inhibitor subunit 16B. Protein phosphatase 1 regulatory inhibitor subunit 16B (PPP1R16B, KIAA0823) is also known as TGF-beta-inhibited membrane-associated protein (TIMAP, hTIMAP), CAAX box protein TIMAP or Ankyrin repeat domain protein 4 (ANKRD4). The protein encoded by this gene is membrane-associated and contains five ankyrin repeats, a protein phosphatase-1-interacting domain, and a carboxy-terminal CAAX box domain.


Synthesis of the encoded protein is inhibited by transforming growth factor beta-1. The protein may bind to the membrane through its CAAX box domain and may act as a signaling molecule through interaction with protein phosphatase-1.


Zinc finger protein SNAI1. Zinc finger protein SNAI1 is also known as Snail protein homolog or Sna protein (SNAI1 alias SNA; SNAH; SLUGH2; dJ710H13.1). The Drosophila embryonic protein snail is a zinc finger transcriptional repressor which downregulates the expression of ectodermal genes within the mesoderm. The nuclear protein encoded by this gene is structurally similar to the Drosophila snail protein, and is also thought to be critical for mesoderm formation in the developing embryo. At least two variants of a similar processed pseudogene have been found on chromosome 2 (Entrez database).


RP11-216C10.1. RP11-216C10.1 is a novel transcript.


XXbac-B444P24.7. XXbac-B444P24.7 also known as Em:AC006547.C22.7 is a novel transcript.


Reticulon 4 receptor precursor. Reticulon 4 receptor precursor is also known as Nogo receptor or Nogo-66 receptor (RTN4R alias NGR; NgR; NOGOR). This gene encodes the receptor for reticulon 4, oligodendrocyte myelin glycoprotein and myelin-associated glycoprotein. This receptor mediates axonal growth inhibition and may play a role in regulating axonal regeneration and plasticity in the adult central nervous system (Entrez database).


NFAT activation molecule 1 precursor. NFAT activation molecule 1 precursor is also known as Calcineurin/NFAT-activating ITAM-containing protein or NFAT activating protein with ITAM motif 1 (NFAM1 alias CNAIP; FLJ40652). The protein encoded by this gene is a type I membrane receptor that activates cytokine gene promoters such as the IL-13 and TNF-alpha promoters. The encoded protein contains an immunoreceptor tyrosine-based activation motif (ITAM) and is thought to regulate the signaling and development of B-cells (Entrez database).


RNA-binding protein Raly. RNA-binding protein Raly (hnRNP associated with lethal yellow homolog) D (RALY alias P542; MGC117312). In infectious mononucleosis, anti-EBNA-1 antibodies are produced which cross-react with multiple normal human proteins. The cross-reactivity is due to anti-gly/ala antibodies that cross-react with host proteins containing configurations like those in the EBNA-1 repeat. One such antigen is RALY which is a member of the heterogeneous nuclear ribonucleoprotein gene family (Entrez database).


Receptor-type tyrosine-protein phosphatase T precursor. Receptor-type tyrosine-protein phosphatase T precursor (EC 3.1.3.48; R-PTP-T alias RPTP-rho; PTPRT; RPTPrho; KIAA0283) also known as protein tyrosine phosphatase receptor type T is a member of the protein tyrosine phosphatase (PTP) family. PTPs are known to be signaling molecules that regulate a variety of cellular processes including cell growth, differentiation, mitotic cycle, and oncogenic transformation. This PTP possesses an extracellular region, a single transmembrane region, and two tandem intracellular catalytic domains, and thus represents a receptor-type PTP. The extracellular region contains a meprin-A5 antigen-PTP (MAM) domain, Ig-like and fibronectin type III-like repeats. The protein domain structure and the expression pattern of the mouse counterpart of this PTP suggest its roles in both signal transduction and cellular adhesion in the central nervous system. Two alternatively spliced transcript variants of this gene, which encode distinct proteins, have been reported (Entrez database).


RP11-191L9.1. RP11-191L9.1 is also known as bA191L9.C22.1.


RP11-410N8.3. RP11-410N8.3 is also known as bA410N8.3.


Lactosylceramide 4-alpha-galactosyltransferase. Lactosylceramide 4-alpha-galactosyltransferase (EC 2.4.1.228; A4GALT alias P1; PK; A14GALT; A4GALT1) catalyzes the transfer of galactose to lactosylceramide to form globotriaosylceramide, which has been identified as the P(k) antigen of the P blood group system. The encoded protein, which is a type II membrane protein found in the Golgi, is also required for the synthesis of the bacterial verotoxins receptor (Entrez database).


Histone H2A. Histone H2A (H1ST1H2AA alias H2AFR; bA317E16.2). Histones are basic nuclear proteins that are responsible for the nucleosome structure of the chromosomal fiber in eukaryotes. Nucleosomes consist of approximately 146 bp of DNA wrapped around a histone octamer composed of pairs of each of the four core histones (H2A, H2B, H3, and H4). The chromatin fiber is further compacted through the interaction of a linker histone, H1, with the DNA between the nucleosomes to form higher order chromatin structures. This gene is intronless and encodes a member of the histone H2A family. Transcripts from this gene contain a palindromic termination element (Entrez database).


Phosphatase and actin regulator 2. Phosphatase and actin regulator 2 is also known as PHACTR2 alias C6orf56; KIAA0680; DKFZp686F18175.


Pannexin-2. Pannexin-2 (PANX2 alias hPANX2; MGC119432) belongs to the innexin family. Innexin family members are the structural components of gap junctions. This protein and pannexin 1 are abundantly expressed in central nerve system (CNS) and are coexpressed in various neuronal populations. Studies in Xenopus oocytes suggest that this protein alone and in combination with pannexin 1 may form cell type-specific gap junctions with distinct properties (Entrez database).


Membrane protein MLC1. Membrane protein MLC1 (MLC1 alias VL; LVM; MLC; KIAA0027) has a sofar unknown function. However, homology to other proteins suggests that it may be an integral membrane transporter. Mutations in this gene have been associated with megalencephalic leukoencephalopathy with subcortical cysts, an autosomal recessive neurological disorder. Two transcript variants encoding the same protein but differing in the 5′ UTR have been noted for this gene (Entrez database).


RP11-318C17.1. RP11-318C17.1 is also known as bA318C17.1.


Immunoglobulin lambda constant 2. Immunoglobulin lambda constant 2 (IGLC2 alias IGLC; MGC20392; MGC45681) is also known as Ig light-chain partial Ke-Oz- polypeptide C-term or immunoglobulin lambda constant region 2 (Kern-Oz- marker).


Potassium voltage-gated channel Shab-related subfamily member 1. Potassium voltage-gated channel, Shab-related subfamily, member 1 (KCNB1 alias DRK1; KV2.1; h-DRK1). Voltage-gated potassium (Kv) channels represent the most complex class of voltage-gated ion channels from both functional and structural standpoints. Their diverse functions include regulating neurotransmitter release, heart rate, insulin secretion, neuronal excitability, epithelial electrolyte transport, smooth muscle contraction, and cell volume. Four sequence-related potassium channel genes—shaker, shaw, shab, and shal—have been identified in Drosophila, and each has been shown to have human homolog(s). This gene encodes a member of the potassium channel, voltage-gated, shab-related subfamily. This member is a delayed rectifier potassium channel and its activity is modulated by some other family members (Entrez database).


Serine/threonine-protein kinase 19. Serine/threonine-protein kinase 19 (EC 2.7.1.37; STK19 alias D6S60; D6S60E; HLA-RP1; MGC117388) is also known as RP1 protein (RP1) or G11 protein (G11). This gene encodes a serine/threonine kinase which localizes predominantly to the nucleus. Its specific function is unknown; it is possible that phosphorylation of this protein is involved in transcriptional regulation. This gene localizes to the major histocompatibility complex (MHC) class III region on chromosome 6 and expresses two transcript variants.


Tubulin alpha-8 chain. Tubulin alpha-8 chain is also known as Alpha-tubulin 8 (TUBA8 alias TUBAL2).


AP000357.3. AP000357.3 is also known as Em:AP000357.C22.3 and might be a pseudogene.


High mobility group protein HMG-I/HMG-Y. High mobility group protein HMG-I/HMG-Y (HMG-I(Y)) is also known as High mobility group AT-hook 1 or High mobility group protein A1 (HMGA1 alias HMG-R; HMGIY; MGC4242; MGC4854; MGC12816). This gene encodes a non-histone protein involved in many cellular processes, including regulation of inducible gene transcription, integration of retroviruses into chromosomes, and the metastatic progression of cancer cells. The encoded protein preferentially binds to the minor groove of A+T-rich regions in double-stranded DNA. It has little secondary structure in solution but assumes distinct conformations when bound to substrates such as DNA or other proteins. The encoded protein is frequently acetylated and is found in the nucleus. At least seven transcript variants encoding two different isoforms have been found for this gene (Entrez database).


Arylsulfatase A precursor. Arylsulfatase A precursor (EC 3.1.6.8; ASA) is also known as Cerebroside-sulfatase (contains: Arylsulfatase A component B; Arylsulfatase A component C) (ARSA alias MLD). The protein encoded by this gene hydrolyzes cerebroside sulfate to cerebroside and sulfate. Defects in this gene lead to metachromatic leucodystrophy (MLD), a progressive demyelination disease which results in a variety of neurological symptoms and ultimately death (Entrez database).


RP11-146I2.2. RP11-146I2.2 is also known as bA146I2.2.


Cold shock domain protein C2. Cold shock domain protein C2 is also known as RNA-binding protein PIPPin (CSDC2_HUMAN alias PIPPIN).


Chromosome 6 open reading frame 190. Chromosome 6 open reading frame 190 is also known as C6orf190; C6orf207; FLJ40584; bA325O24.3 or bA325O24.4.


Metalloproteinase inhibitor 3 precursor. Metalloproteinase inhibitor 3 precursor is also known as Tissue inhibitor of metalloproteinases-3 or MIG-5 protein (TIMP3 alias TIMP-3; SFD; K222; K222TA2; HSMRK222). This gene belongs to the TIMP gene family. The proteins encoded by this gene family are inhibitors of the matrix metalloproteinases, a group of peptidases involved in degradation of the extracellular matrix (ECM). Expression of this gene is induced in response to mitogenic stimulation and this netrin domain-containing protein is localized to the ECM. Mutations in this gene have been associated with the autosomal dominant disorder Sorsby's fundus dystrophy (Entrez database).


Protein C22orf13. Protein C22orf13 is also known as Protein LLN4 or CV013_HUMAN.


HARS2. HARS2 is also known as DUEB; C20orf88; MGC41905; MGC119131; bA379J5.3; bA555E18.1 and is probable a D-tyrosyl-tRNA(Tyr) deacylase (EC 3.1.-.-). The protein encoded by this gene is similar in sequence to histidyl-tRNA synthetase, which hydrolyzes D-tyrosyl-tRNA(Tyr) into D-tyrosine and free tRNA(Tyr). The encoded protein is found in the cytoplasm (Entrez database).


RP4-695O20_B.9. RP4-695O20_B.9 is also known as dJ695O20B.C22.9.


Cat eye syndrome critical region protein 1 precursor. Cat eye syndrome critical region protein 1 precursor (CECR1 alias IDGFL) is member of a subfamily of the adenosine deaminase protein family. The encoded protein may act as a growth factor and have adenosine deaminase activity. It may be responsible for some of the phenotypic features associated with cat eye syndrome. Two transcript variants encoding distinct isoforms have been identified for this gene (Entrez database).


Transcription factor 19. Transcription factor 19 is also known as Transcription factor SC1 (TCF19_HUMAN or SC1).


SUMMARY OF THE INVENTION

The present invention provides novel and efficacious methods and nucleic acids for the classification of biological samples.


The subject matter of the invention has specific utility in the fields of medicine and/or molecular biology. In a particular aspect the utility of the present invention is to provide molecular markers and methods for the analysis thereof that may be considered an alternative to traditional histological or pathological analysis. Said molecular biological markers accordingly offer an alternative to current means such as staining and microscopic analysis.


In a particular aspect, the present invention is of particular use in determining the presence or absence of specific organ, tissue or cell types in a biological sample. Wherein said sample is heterogeneous in nature, the method according to the present invention may be used for the identification of a population or subpopulation of specific organs, tissue or cell types.


In a particular aspect, the present invention has further utility in the detection and/or classification of a cell proliferative disorder, for example but not limited to cancer.


In a particular aspect, the method of the present invention has a further alternative utility in the analysis of cellular differentiation, for example in the field of tissue engineering.


In a particular aspect, the invention solves this longstanding need in the art by providing genes, genomic sequences and/or regulatory regions thereof according to Table 1 (or to one or more of those), the expression thereof being indicative of the class of biological sample.


In a particularly preferred aspect, the methylation status of CpG positions of genes, genomic sequences and/or regulatory regions thereof according to Table 1 is used in the classification of a biological sample.


According to the invention, the provided markers, in particular the genes, genomic sequences, regulatory regions, and corresponding mRNAs, cDNAs, proteins or peptides have a particular utility in the following aspects. Thereby a single marker is used either alone or in combination with other marker or markers herein provided or not.


The herein provided markers have utility (i) for the characterization of the marker corresponding tissue or cell, (ii) for the identification of marker corresponding tissue or cell, (iii) for the isolation of marker corresponding tissue or cell, (iv) for the purification of the corresponding tissue or cell, or (v) combinations thereof. Therefore known methods, so far unreported methods, or combinations thereof are useable. Said application is useful in the field of research, diagnostics as well as therapeutics.


In addition, the herein provided markers have utility for the prospective profiling, retrospective profiling, or both of donors and/or recipients in organ transplantation procedures. The correct characterization, identification, or both of the donor and/or the recipient is mandatory during organ transplantation procedures to assure the success of the intervention. The use of the markers of the invention enables the profiling of both, donor and recipient, form which prospective or retrospective observations or conclusions about the feasibility of the procedure are drawn.


In addition, the herein provided markers have utility for histological, chemical and/or immunohistochemical analysis. Accordingly, they have utility in the fields of research as well as diagnostics, in particular for histological or pathological analysis.


In addition, the herein provided markers have utility for phylogenetic profiling of species or tissues.


The ontogenetic origin or the developmental lineage is then determined by comparison of the determined profiles.


In addition, the herein provided markers have utility for quality control of a genetically modified organism, tissue, group of cells or cell.


In addition, the herein provided markers have utility for controlling side effects in in vivo gene therapy procedures wherein genetically modified organism, tissue, group of cells or cell is used.


In addition, the herein provided markers have utility for the characterization, identification, or labelling of corresponding tissue or combinations thereof. This is of particular utility in the field of tissue bank storage and proliferation. Furthermore it has utility in a prospective as well as in a retrospective manner. The provided markers allow the individualization of samples by a precise molecular method. This is mandatory in storing biological material from patients or healthy individuals. In addition, this also advantageous for isolation or purification of tissues cells.


In addition, the herein provided markers have utility for controlling cell differentiation in stem-cell research and/or therapeutics. Cells undergo many genetic and/or epigenetic changes throughout differentiation. These changes influence the physiology of the cell and their control is mandatory in any procedure involving stem-cell in research and/or therapeutics. The provided markers allow to control this changes by giving a reference of the adult (completely differentiated) and embryonic (partially differentiated) status of the cells.


CD4+ and CD8+ lymphocytes: The herein provided markers of Table 8A and Table 9A have utility for the quantification of lymphocytes, in particular in peripheral blood. The said markers enable the identification of CD4+ and CD8+ lymphocytes among other cells in blood samples. A low number of leucocytes in blood (leucopenia) may indicate bone marrow failure (for example, due to infection, tumor, fibrosis); presence of cytotoxic substance; collagen-vascular diseases (such as lupus erythematosus); disease of the liver or spleen; or exposure to radiation. A high number of leucocytes in blood (leucocytosis) may indicate infectious diseases; inflammatory disease (such as rheumatoid arthritis or allergy); leukemia; severe emotional or physical stress; tissue damage (for example, burns); or anemia.


In addition, the herein provided markers of Table 8A and Table 9A have utility for the study of CD4 and/or CD8 T-lymphocyte infiltration in other tissues healthy or diseased. Infiltration of lymphocytes in healthy or diseased tissues is an indication of several diseases such immunological malignances or even in tumor development. The said markers represent a target for the development of molecular probes that coupled to any detection method (e.g. Fluorescent dye) allow the identification of these cells in histological preparations.


In addition, the herein provided markers of Table 8A and Table 9A have utility for identification, isolation and/or purification of CD4 T-lymphocytes and/or CD8 T-lymphocytes, in particular from surrounding tissue infiltrated by the T-lymphocytes; from blood; and/or from other body fluids.


In addition, the herein provided markers of Table 8A and Table 9A have utility for the identification of an individual. Thereby at least two samples are used. One sample is obtained from an individual and another sample is a forensic sample, in particular traces of body cells, tissues or fluids, for example but not limited to, traces of blood and/or body fluids. This is of particular utility in the field of forensic medicine or of legal medicine. As constituent of blood or body fluids, CD4 T-lymphocytes and CD8 T-lymphocytes are part of the mentioned traces. The said markers have the advantage of being stable over time because they are DNA based. In addition said markers have the advantage that they enable a highly detailed and accurate characterization of samples. Through this an unambiguous identification of an individual is enabled.


In addition, the herein provided markers of Table 8A and Table 9A have utility for diagnosing the presence or absence of a disease. Thereby the number of CD4 T-lymphocytes, CD8 T-lymphocytes or both is quantified in normalized samples of healthy individuals. The determined number of CD4 T-lymphocytes, CD8 T-lymphocytes or both are then considered as indicative for healthy condition or a diseased condition with respect to an individual. Preferably, large amount of normalized samples are considered to generate reference values of CD4 T-lymphocytes, CD8 T-lymphocytes or both for a healthy condition and/or for one or more diseased conditions. The diseased condition can be any kind of diseased condition. Preferably, the diseased condition is a disease which causes a immune reaction. For example but not limited to the diseased condition is a cancer disease, a cell proliferation disease, or HIV invection. Preferably the total number of cells present in a sample is determined. The number of CD4 T-lymphocytes, CD8 T-lymphocytes or both are then normalized to the total number of cells.


Embryonic:

The herein provided markers of Table 8B, Table 8C, Table 9B and Table 9C have utility for the study, identification and/or quantification of fetal cells or fetal DNA circulating in maternal blood and/or amniotic fluid. During pregnancy cells and DNA from the fetus are continuously brought to the maternal blood stream as well as the amniotic fluid. Fetal cells and fetal DNA have a diagnostic potential in monitoring the health status of the fetus as reviewed by Bianchi D, 2004 (Bianchi D W. Circulating fetal DNA: its origin and diagnostic potential-a review. Placenta. 2004 April; 25 Suppl A:S93-S101).


In addition, the herein provided markers of Table 8B, Table 8C, Table 9B and Table 9C have utility for the study, identification and/or quantification of fetal cells or fetal DNA from amniocentesis and/or chorionic villus sampling. This is of particular utility in the field of prenatal diagnosis. Prenatal diagnosis procedures involve the study of fetal cells obtained by amniocentesis and chorionic villus biopsies.


Skin:

The herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility for identifying individuals from traces of skin and/or adjacent tissues (such as hair, nail pieces, etc). This is of particular utility in forensic medicine and/or legal medicine. Skin or skin adjacent tissue is usually used as study material in forensic and legal medicine. The markers provided in Table 8G and 9F have a particular utility because of the following reason: Keratinocytes constitute the external layer of the skin and therefore are the first cell type to be de-attached and a high number of these cells is expected in skin traces. Variations of one marker alone or in combination with other markers herein provided or not enable the accurate assessment of identity.


In addition, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility for characterizing the skin, hair, nail, or adjacent tissue of an individual.


In addition, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility for determining the composition of the skin, hair, nail, or adjacent tissue of an individual. Said composition being dependent from the content of at least one of the three major constituting cell types of the skin (fibroblasts, keratinocytes and melanocytes).


In addition, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility in the field of drugs. They have particular utility for the development of drugs as well as for the treatment with drugs. The skin, hair, nail or adjacent tissue of an individual can be characterized by means of the provided markers of Tables 8D, G, I and Tables 9D, F, H. This information can then be used to develop new drugs or to access already existing drugs with regard to skin, hair, nail etc. of an individual or to subgroups of individuals. These subgroups are for example but not limited to be characterized by a disease and/or a defined type of skin or hair, etc. The efficiency of said drugs i.e. the presence or absence of the desired effect is also characterized or monitored by means of the provided markers of Tables 8D, G, I and Tables 9D, F, H.


In addition, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility as prognostic and/or diagnostic markers for wound healing, in particular in the field of surgery procedures wherein the skin is affected.


Liver:

The herein provided markers of Tables 8H and Tables 9G have utility for deducing the presence of absence of an event which affects the liver. For example but not limited to it, said event is at least one select from the group comprising liver cirrhosis; liver cancer; hepatitis A; hepatitis B; hepatitis C; healthy condition, recently or longer chemical, physical or biological exposure; recently or longer exposure to a drug, or alcohol; or treatment procedures. In the case the event is adverse, said event affecting the liver leads to the death of liver cells. In the case the event is benign, said event leads to a reduction of liver cell death. The genomic DNA of dead liver cells can then be found in the body fluids in particular in the blood of a affected individual.


In addition, the herein provided markers of Tables 8H and Tables 9G have utility for deducing the sensitivity of an individual to alcohol. Alcohol consumption may change the DNA methylation status as reviewed by Poschl et al, 2004 (Poschl G, Stickel F, Wang X D, Seitz H K. Alcohol and cancer: genetic and nutritional aspects. Proc Nutr Soc. 2004 February; 63(1):65-71.).


Heart Muscle:

The herein provided markers of Tables 8E, Table 8F and Tables 9E have utility for deducing the presence of absence of an event or condition affecting the heart. For example but not limited to it, said event or condition is at least one select from the group comprising heart failure; heart attack; athletic capacity; healthy condition; recently or longer chemical, physical or biological exposure; recently or longer exposure to a drug; or treatment procedure. In the case the event is adverse, said event or condition affecting the heart leads to death of heart cells. In the case the event is benign, said event leads to a reduction of heart cell death. The genomic DNA of dead heart cells can then be found in the body fluids in particular in the blood of an affected individual.


Placenta:

The herein provided markers of Table 8J and Table 9I have utility for the study, monitoring, identification and/or quantification of placental cells or placental DNA circulating in maternal blood and/or amniotic fluid. In this respect, the said markers have also utility for the isolation or purification of placental cell or placental genomic DNA. Placenta constitute an extra-embryonic fetal tissue and as such, it shares many genetic characteristics with the fetal tissue. Therefore, cells from the placenta as well as DNA from placental cells can surrogate fetal cells and fetal DNA for diagnostic means. Fetal cells and fetal DNA have a diagnostic potential in monitoring the health status of the fetus as reviewed by Bianchi D, 2004 (Bianchi D W. Circulating fetal DNA: its origin and diagnostic potential-a review. Placenta. 2004 April; 25 Suppl A:S93-S101). During pregnancy placenta cells are de-attached and brought to the maternal blood stream as well as the amniotic fluid.


In addition, the herein provided markers of Table 8J and Table 9I have utility for the monitoring of embryonic development or the monitoring of placental development, in particular of extra-embryonic tissue or of interaction of extra-embryonic tissue with maternal placental tissue.


In addition, the herein provided markers of Table 8J and Table 9I have utility for the study, monitoring, identification and/or quantification of placental cells in regenerative medicine, in particular in the field of tissue engineering. Corresponding methods for the study, monitoring, identification and/or quantification of placental cells are applied in particular before and after storage, before and after cell differentiation, before and after cell proliferation, before and after cell culture expansion, and before and after tissue expansion as well as before and after transplantation.


Sperm:

The herein provided markers of Table 8L have utility for diagnosing a male infertility related disease. A major cause of male infertility is either a low amount of sperm cells (spermatozoa) in the ejaculate (oligospermia) or a complete lack of sperm cells (spermatozoa) in the ejaculate (azoospermia). Thus, methods for the quantification of sperm cells are widely useable in diagnosing male infertility.


In addition, the herein provided markers of Table 8L have utility as a tool to access the viability of the sperm cells.


In addition, the herein provided markers of Table 8L have utility for increasing the fertility of a male individual. As said above male fertility is often limited by the amount of sperm cells in the ejaculate. Thus, male fertility can be enhanced by enriching, isolating or purifying sperm cells.


In addition, the herein provided markers of Table 8L have utility for assisted fertilization procedures. Assisted fertilization procedures are for example but not limited to intracytoplasmic sperm injection (ICSI) or in vitro fertilization (IVF). All assisted fertilization procedures require the management of sperm cells prior to the procedure. Such management comprises at least the characterization, identification, quantification, enrichment, isolation, purification of sperm cells or combinations thereof.


In addition, the herein provided markers of Table 8L have utility in the fields of forensic and/or legal medicine. By use of the said markers it is possible to determine the presence or absence of sperm in a sample. Furthermore, it is possible to identify an individual by use of said markers.


Skeletal Muscle:

The herein provided markers of Table 8F, 8K and Table 9J have utility for characterizing the efficiency of skeletal muscle cells. This utility is of particular value in the field of sports medicine.


In addition, the herein provided markers of Table 8F, 8K and Table 9J have utility for identifying fully differentiated muscle cells in cell culture. This is of particular utility in the field of tissue engineering. Muscle cells are generate in cell culture by cultivation and differentiation of muscle cell progenitor cells. Fully differentiated skeletal muscle cells can be identified by means of the provided markers of Table 8F, 8K and Table 9J.


In addition, the herein provided markers of Table 8F, 8K and Table 9J have utility for diagnosing muscle cell associated diseases, in particular disease which are characterized by a death of muscle cells like muscular distrophy. The DNA of dead muscle cells is found in body fluids such as blood or urine. This DNA can be identified by means of the herein provided markers of Table 8F, 8K and Table 9J.


CD8 T-Lymphocytes:

The herein provided markers of Table 8A specific only for CD8 T-lymphocytes have utility for quantifying CD8 T-lymphocytes, in particular for monitoring the immune system of individuals infected with HIV. The periodically determining of the number of CD8 T-lymphocytes for patients infected with HIV is a standard procedure in the art. It is necessary to decide whether and when a drug or treatment is necessary, whether a drug or treatment is still effective, and which drug or treatment can be selected. The said is necessary with respect to the HIV infection itself but also with respect to secondary infection.


CD4+ Lymphocytes:

The herein provided markers of Table 8A specific only for CD4 T-lymphocytes have utility for quantifying CD4 T-lymphocytes, in particular for monitoring the immune system of individuals infected with HIV. The periodically determining of the number of CD4 T-lymphocytes for patients infected with HIV is a standard procedure in the art. It is necessary to decide whether and when a drug or treatment is necessary, whether a drug or treatment is still effective, and which drug or treatment can be selected. The said is necessary with respect to the HIV infection itself but also with respect to secondary infection.


It is particularly preferred that said biological sample is classified according to at least one parameter selected from the group consisting of the cell, organ or tissue type of said biological sample or features thereof such as disease state.


To enable this analysis the invention provides a method for the analysis of biological samples for genomic methylation associated with the classification of biological samples. Said method is characterized in that at least one nucleic acid, or a fragment thereof, from the group consisting of SEQ ID NO: 413 to SEQ ID NO: 824 is/are contacted with a reagent or series of reagents capable of distinguishing between methylated and non methylated CpG dinucleotides within the genomic sequence, or sequences of interest.


It is particularly preferred that the method and nucleic acids according to the invention are utilised for at least one of histological analysis, pathological analysis, detection and/or characterization of cell proliferative disorders and monitoring of cellular or tissue differentiation.


The DNA source may be any suitable source. Preferably, the source of the DNA sample is selected from the group consisting of cells or cell lines, histological slides, biopsies, surgical samples, tissue samples, body fluids, sputum, stool, nipple aspirate, cerebrospinal fluid, ejaculate, urine, serum, plasma, whole blood, saliva, fluids from the pleural or peritoneal cavity, cerebrospinal fluid or a smear from a epithelial surface and combinations thereof.


Furthermore, said sample may be fresh or archived and can be treated by any means standard in the art, for example but not limited to fresh-frozen, paraffin-embedded or formalin-fixed sample.


Specifically, the present invention provides a method for classifying a biological sample, comprising: obtaining a biological sample comprising genomic nucleic acid(s); contacting the nucleic acid(s), or a fragment thereof, with one reagent or a plurality of reagents sufficient for distinguishing between methylated and non methylated CpG dinucleotide sequences within a target sequence of the subject nucleic acid, wherein the target sequence comprises, or hybridizes under stringent conditions to, a sequence comprising at least 16 contiguous nucleotides of a gene or genomic sequence selected from Table 1 said contiguous nucleotides comprising at least one CpG dinucleotide sequence; and determining, based at least in part on said distinguishing, the methylation state of at least one target CpG dinucleotide sequence, or an average, or a value reflecting an average methylation state of a plurality of target CpG dinucleotide sequences. Preferably said target sequences are selected from Table 3, which provides particularly preferred regions of the sequences of Table 1. Preferably, distinguishing between methylated and non methylated CpG dinucleotide sequences within the target sequence comprises methylation state-dependent conversion or non-conversion of at least one such CpG dinucleotide sequence to the corresponding converted or non-converted dinucleotide sequence within a sequence selected from the equivalent converted sequence selected from Table 1, and contiguous regions thereof corresponding to the target sequence. It is further preferred that said converted sequences are selected from Table 3, which provides particularly preferred regions of the genes according to Table 1 (converted sequences thereof provided in Table 2).


Additional embodiments provide a method for the classification of a biological sample, comprising: obtaining a biological sample having subject genomic DNA; extracting the genomic DNA; treating the genomic DNA, or a fragment thereof, with one or more reagents to convert 5-position unmethylated cytosine bases to uracil or to another base that is detectably dissimilar to cytosine in terms of hybridization properties; contacting the treated genomic DNA, or the treated fragment thereof, with an amplification enzyme and at least two primers comprising, in each case a contiguous sequence at least 9 nucleotides in length that is complementary to, or hybridizes under moderately stringent or stringent conditions to a converted sequence selected from Table 1 and complements thereof, wherein the treated DNA or the fragment thereof is either amplified to produce an amplificate, or is not amplified; and determining, based on a presence or absence of, or on a property of said amplificate, the methylation state of at least one CpG dinucleotide sequence of a gene or genomic sequence selected form Table 1, or an average, or a value reflecting an average methylation state of a plurality of CpG dinucleotide sequences thereof.


Preferably, determining comprises use of at least one method selected from the group consisting of: hybridizing at least one nucleic acid molecule comprising a contiguous sequence at least 9 nucleotides in length that is complementary to, or hybridizes under moderately stringent or stringent conditions to a converted sequence selected from Table 2 and complements thereof, hybridizing at least one nucleic acid molecule, bound to a solid phase, comprising a contiguous sequence at least 9 nucleotides in length that is complementary to, or hybridizes under moderately stringent or stringent conditions to a converted sequence selected from Table 2, and complements thereof; hybridizing at least one nucleic acid molecule comprising a contiguous sequence at least 9 nucleotides in length that is complementary to, or hybridizes under moderately stringent or stringent conditions to a sequence selected from a converted sequence selected from Table 2 (SEQ ID NO: 1650 to SEQ ID NO: 4120), and complements thereof, and extending at least one such hybridized nucleic acid molecule by at least one nucleotide base; and sequencing of the amplificate.


Further embodiments provide a method for classifying a biological sample, comprising: obtaining a biological sample having subject genomic DNA; extracting the genomic DNA; contacting the genomic DNA, or a fragment thereof, comprising one or more sequences selected from the group consisting of SEQ ID NO: 413 to SEQ ID NO: 824 or a sequence that hybridizes under stringent conditions thereto, with one or more methylation-sensitive restriction enzymes, wherein the genomic DNA is either digested thereby to produce digestion fragments, or is not digested thereby; and determining, based on a presence or absence of, or on property of at least one such fragment, the methylation state of at least one CpG dinucleotide sequence of one or more sequences selected from the group consisting of SEQ ID NO: 413 to SEQ ID NO: 824, or an average, or a value reflecting an average methylation state of a plurality of CpG dinucleotide sequences thereof. Preferably, the digested or undigested genomic DNA is amplified prior to said determining.


Additional embodiments provide novel genomic and chemically modified nucleic acid sequences, as well as oligonucleotides and/or PNA-oligomers for analysis of cytosine methylation patterns within sequences from the group consisting of SEQ ID NO: 413 to SEQ ID NO: 824.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1.1 to 1.403 provide the results of bisulphite sequencing as carried out in Example 2. Each individual matrix represents the sequencing data for an individual amplificate. Each of the discrete blocks of the matrix represent a single sample type and are labeled ‘A’ through ‘L’, said letters representing in each case the following tissue/cell types: A: Melanocytes; B: Heart Muscle: C: Skeletal muscle; D: Liver; E: Sperm; F: Embryonic skeletal muscle; G: Embryonic liver; H: Placental; I: Fibroblast; J: Keratinocytes; K: CD8; L: CD4. The SEQ ID NO: of the genomic region of each amplificate is shown to the left of the matrices. This may be cross referenced in Table 4 to determine the amplificate and primer sequences. Each row of a matrix represents a single CpG site within the amplificate (according to the corresponding SEQ ID NO: from Table 4) and is numbered accordingly, each column represents a single pooled DNA sample. The degree of methylation is represented by the shade of each position within the column from black representing 100% methylation to light gray representing 0% methylation. White positions represented a measurement for which no data was available.



FIG. 2: Example of RT-PCR results. Studied samples are arranged in columns and genes in rows. High DNA methylation correlates with gene silencing in MYO18B, SLC22A1 and PLG. SERPINB5 expression has been previously reported as silenced by DNA methylation (Futscher et al, 2002). ACTIN B1 is a housekeeping gene and its expression in all analyzed samples confirms the feasibility of the assay. + Control is the amplification of cDNA produced from a pool of total RNAs from several tissues. − RT is a negative control, where no reverse transcriptase was added to the cDNA synthesis reaction.





DETAILED DESCRIPTION OF THE INVENTION
Definitions

As used herein the term “classification” shall be taken to mean the action or process of categorising an object according to pre-determined parameters. Said categorization may be performed by humans means or by means of a computer or computer implemented means. It is particularly preferred that said parameters are phenotypic parameters, accordingly it is particularly preferred that said categorization is the assignment of a biological sample to a particular phenotypic class. In one embodiment of the method said phenotypic parameter or class is selected from the group consisting cell type, organ type, tissue type and disease status.


As used herein the term “expression” shall be taken to mean the transcription and translation of a gene. The level of expression of a gene may be determined by the analysis of any factors associated with or indicative of the level of transcription and translation of a gene including but not limited to methylation analysis, loss of heterozygosity (hereinafter also referred to as LOH), RNA expression levels and protein expression levels.


Furthermore the activity of the transcribed gene may be affected by genetic variations such as but not limited genetic mutations (including but not limited to SNPs, point mutations, deletions, insertions, repeat length, rearrangements and other polymorphisms).


The term “Observed/Expected Ratio” (“O/E Ratio”) refers to the frequency of CpG dinucleotides within a particular DNA sequence, and corresponds to the [number of CpG sites/(number of C bases×number of G bases)].


The term “CpG island” refers to a contiguous region of genomic DNA that satisfies the criteria of (1) having a frequency of CpG dinucleotides corresponding to an “Observed/Expected Ratio” >0.6, and (2) having a “GC Content” >0.5. CpG islands are typically, but not always, between about 0.2 to about 1 kb, or to about 2 kb in length.


The term “methylation state” or “methylation status” refers to the presence or absence of 5-methylcytosine (“5-mCyt”) at one or a plurality of CpG dinucleotides within a DNA sequence. Methylation states at one or more particular CpG methylation sites (each having two CpG dinucleotide sequences) within a DNA sequence include “unmethylated,” “fully-methylated” and “hemi-methylated.”


The term “methylation level” or “level of methylation” refers to the degree of 5-methylcytosine (“5-mCyt”) at one or a plurality of CpG dinucleotides within a DNA sequence wherein one or more DNA molecules are considered.


The term “hemi-methylation” or “hemimethylation” refers to the methylation state of a palindromic CpG methylation site, where only a single cytosine in one of the two CpG dinucleotide sequences of the palindromic CpG methylation site is methylated (e.g., 5′-CCMGG-3′ (top strand): 3′-GGCC-5′ (bottom strand)).


The term ‘AUC’ as used herein is an abbreviation for the area under a curve. In particular it refers to the area under a Receiver Operating Characteristic (ROC) curve. The ROC curve is a plot of the true positive rate against the false positive rate for the different possible cutpoints of a diagnostic test. It shows the tradeoff between sensitivity and specificity depending on the selected cutpoint (any increase in sensitivity will be accompanied by a decrease in specificity). The area under an ROC curve (AUC) is a measure for the accuracy of a diagnostic test (the larger the area the better, optimum is 1, a random test would have a ROC curve lying on the diagonal with an area of 0.5; for reference: J. P. Egan. Signal Detection Theory and ROC Analysis, Academic Press, New York, 1975). The term “hypermethylation” refers to the average methylation state corresponding to an increased presence of 5-mCyt at one or a plurality of CpG dinucleotides within a DNA sequence of a test DNA sample, relative to the amount of 5-mCyt found at corresponding CpG dinucleotides within a normal control DNA sample.


The term “hypomethylation” refers to the average methylation state corresponding to a decreased presence of 5-mCyt at one or a plurality of CpG dinucleotides within a DNA sequence of a test DNA sample, relative to the amount of 5-mCyt found at corresponding CpG dinucleotides within a normal control DNA sample.


The term “microarray” refers broadly to both “DNA microarrays,” and ‘DNA chip(s),’ as recognized in the art, encompasses all art-recognized solid supports, and encompasses all methods for affixing nucleic acid molecules thereto or synthesis of nucleic acids thereon.


“Genetic parameters” are mutations and polymorphisms of genes and sequences further required for their regulation. To be designated as mutations are, in particular, insertions, deletions, point mutations, inversions and polymorphisms and, particularly preferred, SNPs (single nucleotide polymorphisms).


“Epigenetic parameters” are, in particular, cytosine methylations. Further epigenetic parameters include, for example, the acetylation of histones which, however, cannot be directly analyzed using the described method but which, in turn, correlate with the DNA methylation.


The term “bisulfite reagent” refers to a reagent comprising bisulfite, disulfite, hydrogen sulfite or combinations thereof, useful as disclosed herein to distinguish between methylated and unmethylated CpG dinucleotide sequences.


The term “Methylation assay” refers to any assay for determining the methylation state of one or more CpG dinucleotide sequences within a sequence of DNA.


The term “MS.AP-PCR” (Methylation-Sensitive Arbitrarily-Primed Polymerase Chain Reaction) refers to the art-recognized technology that allows for a global scan of the genome using CG-rich primers to focus on the regions most likely to contain CpG dinucleotides, and described by Gonzalgo et al., Cancer Research 57:594-599, 1997.


The term “MethyLight™” refers to the art-recognized fluorescence-based real-time PCR technique described by Eads et al., Cancer Res. 59:2302-2306, 1999.


The term “HeavyMethyl™” assay, in the embodiment thereof implemented herein, refers to an assay, wherein methylation specific blocking probes (also referred to herein as blockers) covering CpG positions between, or covered by the amplification primers enable methylation-specific selective amplification of a nucleic acid sample.


The term “HeavyMethyl™ MethyLight™” assay, in the embodiment thereof implemented herein, refers to a HeavyMethyl™ MethyLight™ assay, which is a variation of the MethyLight™ assay, wherein the MethyLight™ assay is combined with methylation specific blocking probes covering CpG positions between the amplification primers.


The term “Ms-SNuPE” (Methylation-sensitive Single Nucleotide Primer Extension) refers to the art-recognized assay described by Gonzalgo and Jones, Nucleic Acids Res. 25:2529-2531, 1997.


The term “MSP” (Methylation-specific PCR) refers to the art-recognized methylation assay described by Herman et al. Proc. Natl. Acad. Sci. USA 93:9821-9826, 1996, and by U.S. Pat. No. 5,786,146.


The term “COBRA” (Combined Bisulfite Restriction Analysis) refers to the art-recognized methylation assay described by Xiong and Laird, Nucleic Acids Res. 25:2532-2534, 1997.


The term “MCA” (Methylated CpG Island Amplification) refers to the methylation assay described by Toyota et al., Cancer Res. 59:2307-12, 1999, and in WO 00/26401A1.


The term “hybridization” is to be understood as a bond of an oligonucleotide to a complementary sequence along the lines of the Watson-Crick base pairings in the sample DNA, forming a duplex structure.


“Stringent hybridization conditions,” as defined herein, involve hybridizing at 68° C. in 5×SSC/5×Denhardt's solution/1.0% SDS, and washing in 0.2×SSC/0.1% SDS at room temperature, or involve the art-recognized equivalent thereof (e.g., conditions in which a hybridization is carried out at 60° C. in 2.5×SSC buffer, followed by several washing steps at 37° C. in a low buffer concentration, and remains stable). Moderately stringent conditions, as defined herein, involve including washing in 3×SSC at 42° C., or the art-recognized equivalent thereof. The parameters of salt concentration and temperature can be varied to achieve the optimal level of identity between the probe and the target nucleic acid. Guidance regarding such conditions is available in the art, for example, by Sambrook et al., 1989, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, N.Y.; and Ausubel et al. (eds.), 1995, Current Protocols in Molecular Biology, (John Wiley and Sons, N.Y.) at Unit 2.10.


The term polypeptide as used hereunder shall be taken to encompass all peptides, proteins and/or fragments thereof.


Overview:

The present invention provides for molecular genetic markers that have novel utility for the classification of biological samples. In particular embodiments classification is according to at least one parameter selected from the group consisting cell type, organ type, tissue type and disease status. It is particularly preferred that the method and nucleic acids according to the invention are utilized for at least one of histological analysis, pathological analysis, detection and/or characterization of cell proliferative disorders and monitoring of cellular or tissue differentiation.


In a particularly preferred embodiment the invention provides a method for the classification of biological samples, comprising the following steps:


a) determining the expression levels of one or more genes or gene sequences according to Table 1 and/or regulatory regions thereof; and


b) classifying said biological sample according to said expression status. Said expression level may be determined by any means standard in the art including but not limited to methylation analysis, loss of heterozygosity (hereinafter also referred to as LOH), RNA expression levels and protein expression levels.


Accordingly, said method may be enabled by means of any analysis of the expression of a RNA transcribed therefrom or polypeptide or protein translated from said RNA, preferably by means of mRNA expression analysis or polypeptide expression analysis. Accordingly the present invention also provides classification assays and methods, both quantitative and qualitative for detecting the expression of the genes, genomic sequences and/or regulatory regions according to Table 1 and providing therefrom a classification of said biological sample.


Expression of mRNA transcribed from the genes or genomic regions according to Table 1, are associated with specific organ and cell types.


To detect the presence of mRNA encoding a gene or genomic sequence, a sample is obtained from a patient. The sample may be any suitable sample comprising cellular matter of the tumor, most preferably the primary tumor. Suitable sample types include cells or cell lines, histological slides, biopsies, paraffin-embedded tissue, bodily fluids, sputum, stool, nipple aspirate, cerebrospinal fluid, ejaculate, urine, blood or any other suitable biological sample and all possible combinations thereof.


The sample may be treated to extract the RNA contained therein. The resulting nucleic acid from the sample is then analyzed. Many techniques are known in the state of the art for determining absolute and relative levels of gene expression, commonly used techniques suitable for use in the present invention include in situ hybridization (e.g. FISH), Northern analysis, RNase protection assays (RPA), microarrays and PCR-based techniques, such as quantitative PCR and differential display PCR or any other nucleic acid detection method.


Particularly preferred is the use of the reverse transcription/polymerisation chain reaction technique (RT-PCR). The method of RT-PCR is well known in the art (for example, see Watson and Fleming, supra).


The RT-PCR method can be performed as follows. Total cellular RNA is isolated by, for example, the standard guanidium isothiocyanate method and the total RNA is reverse transcribed. The reverse transcription method involves synthesis of DNA on a template of RNA using a reverse transcriptase enzyme and a 3′ end oligo dT primer and/or random hexamer primers. The cDNA thus produced is then amplified by means of PCR. (Belyavsky et al, Nucl Acid Res 17:2919-2932, 1989; Krug and Berger, Methods in Enzymology, Academic Press, N.Y., Vol. 152, pp. 316-325, 1987 which are incorporated by reference). Further preferred is the “Real-time” variant of RT-PCR, wherein the PCR product is detected by means of hybridization probes (e.g. TaqMan, Lightcycler, Molecular Beacons and Scorpion) or SYBR green. The detected signal from the probes or SYBR green is then quantified either by reference to a standard curve or by comparing the Ct values to that of a calibration standard. Analysis of housekeeping genes is often used to normalize the results.


In Northern blot analysis total or poly(A)+ mRNA is run on a denaturing agarose gel and detected by hybridization to a labeled probe in the dried gel itself or on a membrane. The resulting signal is proportional to the amount of target RNA in the RNA population.


Comparing the signals from two or more cell populations or tissues reveals relative differences in gene expression levels. Absolute quantification can be performed by comparing the signal to a standard curve generated using known amounts of an in vitro transcript corresponding to the target RNA. Analysis of housekeeping genes, genes whose expression levels are expected to remain relatively constant regardless of conditions, is often used to normalize the results, eliminating any apparent differences caused by unequal transfer of RNA to the membrane or unequal loading of RNA on the gel.


The first step in Northern analysis is isolating pure, intact RNA from the cells or tissue of interest. Because Northern blots distinguish RNAs by size, sample integrity influences the degree to which a signal is localized in a single band. Partially degraded RNA samples will result in the signal being smeared or distributed over several bands with an overall loss in sensitivity and possibly an erroneous interpretation of the data. In Northern blot analysis, DNA, RNA and oligonucleotide probes can be used and these probes are preferably labeled (e.g. radioactive labels, mass labels or fluorescent labels). The size of the target RNA, not the probe, will determine the size of the detected band, so methods such as random-primed labeling, which generates probes of variable lengths, are suitable for probe synthesis. The specific activity of the probe will determine the level of sensitivity, so it is preferred that probes with high specific activities, are used.


In an RNase protection assay, the RNA target and an RNA probe of a defined length are hybridized in solution. Following hybridization, the RNA is digested with RNases specific for single-stranded nucleic acids to remove any unhybridized, single-stranded target RNA and probe. The RNases are inactivated, and the RNA is separated e.g. by denaturing polyacrylamide gel electrophoresis. The amount of intact RNA probe is proportional to the amount of target RNA in the RNA population. RPA can be used for relative and absolute quantification of gene expression and also for mapping RNA structure, such as intron/exon boundaries and transcription start sites. The RNase protection assay is preferable to Northern blot analysis as it generally has a lower limit of detection.


The antisense RNA probes used in RPA are generated by in vitro transcription of a DNA template with a defined endpoint and are typically in the range of 50-600 nucleotides. The use of RNA probes that include additional sequences not homologous to the target RNA allows the protected fragment to be distinguished from the full-length probe. RNA probes are typically used instead of DNA probes due to the ease of generating single-stranded RNA probes and the reproducibility and reliability of RNA:RNA duplex digestion with RNases (Ausubel et al. 2003), particularly preferred are probes with high specific activities.


Particularly preferred is the use of microarrays. The microarray analysis process can be divided into two main parts. First is the immobilization of known gene sequences onto glass slides or other solid support followed by hybridization of the fluorescently labeled cDNA (comprising the sequences to be interrogated) to the known genes immobilized on the glass slide. After hybridization, arrays are scanned using a fluorescent microarray scanner. Analyzing the relative fluorescent intensity of different genes provides a measure of the differences in gene expression.


DNA arrays can be generated by immobilizing presynthesized oligomers onto prepared glass slides. In this case, representative gene sequences are manufactured and prepared using standard oligomer synthesis and purification methods. These synthesized gene sequences are complementary to the genes of interest and tend to be shorter sequences in the range of 25-70 nucleotides. Alternatively, immobilized oligomers can be chemically synthesized in-situ on the surface of the slide. In situ oligomer synthesis involves the consecutive addition of the appropriate nucleotides to the spots on the microarray; spots not receiving a nucleotide are protected during each stage of the process using physical or virtual masks.


In expression profiling microarray experiments, the RNA templates used are representative of the transcription profile of the cells or tissues under study. RNA is first isolated from the cell populations or tissues to be compared. Each RNA sample is then used as a template to generate fluorescently labeled cDNA via a reverse transcription reaction. Fluorescent labeling of the cDNA can be accomplished by either direct labeling or indirect labeling methods. During direct labeling, fluorescently modified nucleotides (e.g., Cy®3- or Cy®5-dCTP) are incorporated directly into the cDNA during the reverse transcription. Alternatively, indirect labeling can be achieved by incorporating aminoallyl-modified nucleotides during cDNA synthesis and then conjugating an N-hydroxysuccinimide (NHS)-ester dye to the aminoallyl-modified cDNA after the reverse transcription reaction is complete. Alternatively, the probe may be unlabeled, but may be detectable by specific binding with a ligand which is labeled, either directly or indirectly. Suitable labels and methods for labelling ligands (and probes) are known in the art, and include, for example, radioactive labels which may be incorporated by known methods (e.g., nick translation or kinasing). Other suitable labels include but are not limited to biotin, fluorescent groups, chemiluminescent groups (e.g., dioxetanes, particularly triggered dioxetanes), enzymes, antibodies, and the like.


To perform differential gene expression analysis, cDNA generated from different RNA samples are labeled with Cy®3. The resulting labeled cDNA is purified to remove unincorporated nucleotides, free dye and residual RNA. Following purification, the labeled cDNA samples are hybridized to the microarray. The stringency of hybridization is determined by a number of factors during hybridization and during the washing procedure, including temperature, ionic strength, length of time and concentration of fromamide. These factors are outlined in, for example, Sambrook et al. (Molecular Cloning: A Laboratory Manual, 2nd ed., 1989). The microarray is scanned post-hybridization using a fluorescent microarray scanner. The fluorescent intensity of each spot indicates the level of expression for that gene; bright spots correspond to strongly expressed genes, while dim spots indicate weak expression.


Once the images are obtained, the raw data must be analyzed. First, the background fluorescence must be subtracted from the fluorescence of each spot. The data is then normalized to a control sequence, such as an exogenously added RNA, or a housekeeping gene panel to account for any nonspecific hybridization, array imperfections or variability in the array setup, cDNA labeling, hybridization or washing. Data normalization allows the results of multiple arrays to be compared.


The present invention further provides methods for the detection of the presence of the polypeptide encoded by said gene sequences in a sample obtained from a patient.


Levels of polypeptide expression of the polypeptides encoded by the genes and/or genomic regions according to Table 1 are associated with the classification of biological samples.


Any method known in the art for detecting polypeptides can be used. Such methods include, but are not limited to mass-spectrometry, immunodiffusion, immunoelectrophoresis, immunochemical methods, binder-ligand assays, immunohistochemical techniques, agglutination and complement assays (e.g., see Basic and Clinical Immunology, Sites and Terr, eds., Appleton and Lange, Norwalk, Conn. pp 217-262, 1991 which is incorporated by reference). Preferred are binder-ligand immunoassay methods including reacting antibodies with an epitope or epitopes and competitively displacing a labeled polypeptide or derivative thereof.


Certain embodiments of the present invention comprise the use of antibodies specific to the polypeptide encoded by the genes and/or genomic regions according to Table 1.


Such antibodies are useful for the classification of biological samples. In certain embodiments production of monoclonal or polyclonal antibodies can be induced by the use of the coded polypeptide as an antigene. Such antibodies may in turn be used to detect expressed polypeptides as classification markers for biological samples. The levels of such polypeptides present may be quantified by conventional methods. Antibody-polypeptide binding may be detected and quantified by a variety of means known in the art, such as labelling with fluorescent or radioactive ligands. The invention further comprises kits for performing the above-mentioned procedures, wherein such kits contain antibodies specific for the investigated polypeptides.


Numerous competitive and non-competitive polypeptide binding immunoassays are well known in the art. Antibodies employed in such assays may be unlabeled, for example as used in agglutination tests, or labeled for use a wide variety of assay methods. Labels that can be used include radionuclides, enzymes, fluorescers, chemiluminescers, enzyme substrates or co-factors, enzyme inhibitors, particles, dyes and the like. Preferred assays include but are not limited to radioimmunoassay (RIA), enzyme immunoassays, e.g., enzyme-linked immunosorbent assay (ELISA), fluorescent immunoassays and the like. Polyclonal or monoclonal antibodies or epitopes thereof can be made for use in immunoassays by any of a number of methods known in the art.


In an alternative embodiment of the method the proteins may be detected by means of western blot analysis. Said analysis is standard in the art, briefly proteins are separated by means of electrophoresis e.g. SDS-PAGE. The separated proteins are then transferred to a suitable membrane (or paper) e.g. nitrocellulose, retaining the spacial separation achieved by electrophoresis. The membrane is then incubated with a generic protein (e.g. milk protein) to bind remaining sticky places on the membrane. An antibody specific to the protein of interest is then added, said antibody being detectably labeled for example by dyes or enzymatic means (e.g. alkaline phosphatase or horseradish peroxidase). The location of the antibody on the membrane is then detected.


In an alternative embodiment of the method the proteins may be detected by means of immunochemistry (the use of antibodies to probe specific antigens in a sample). Said analysis is standard in the art, wherein detection of antigens in tissues is known as immunohistochemistry, while detection in cultured cells is generally termed immunocytochemistry. Briefly the primary antibody to be detected by binding to its specific antigen. The antibody-antigen complex is then bound by a secondary enzyme conjugated antibody. In the presence of the necessary substrate and chromogen the bound enzyme is detected according to colored deposits at the antibody-antigen binding sites. There is a wide range of suitable sample types, antigen-antibody affinity, antibody types, and detection enhancement methods. Thus optimal conditions for immunochemical detection must be determined by the person skilled in the art for each individual case.


One approach for preparing antibodies to a polypeptide is the selection and preparation of an amino acid sequence of all or part of the polypeptide, chemically synthesising the amino acid sequence and injecting it into an appropriate animal, usually a rabbit or a mouse (Milstein and Kohler Nature 256:495-497, 1975; Gulfre and Milstein, Methods in Enzymology: Immunochemical Techniques 73:1-46, Langone and Banatis eds., Academic Press, 1981 which are incorporated by reference). Methods for preparation of the polypeptides or epitopes thereof include, but are not limited to chemical synthesis, recombinant DNA techniques or isolation from biological samples.


In the final step of the method the biological sample is classified, as specified below.


Another aspect of the invention provides a kit for use in classifying a biological sample, comprising: a means for detecting polypeptides of a gene or genomic region according to Table 1. The means for detecting the polypeptides comprise preferably antibodies, antibody derivatives, or antibody fragments. The polypeptides are most preferably detected by means of Western blotting utilizing a labeled antibody. In another embodiment of the invention the kit further comprising means for obtaining a biological sample of the patient. Preferred is a kit, which further comprises a container suitable for containing the means for detecting the polypeptides in the biological sample of the patient, and most preferably further comprises instructions for use and interpretation of the kit results. In a preferred embodiment the kit for use in classifying biological samples, comprises: (a) a means for detecting polypeptides of a gene or genomic region according to Table 1; (b) a container suitable for containing the said means and the biological sample of the patient comprising the polypeptides wherein the means can form complexes with the polypeptides; (c) a means to detect the complexes of (b); and optionally (d) instructions for use and interpretation of the kit results.


The kit may also contain other components such as buffers or solutions suitable for blocking, washing or coating, packaged in a separate container.


Another aspect of the invention relates to a kit for use in classifying a biological sample, said kit comprising: a means for measuring the level of transcription of a gene or genomic region according to Table 1. In a preferred embodiment the means for measuring the level of transcription comprise oligomers or polynucleotides able to hybridize under stringent or moderately stringent conditions to the transcription products of a gene or genomic region according to Table 1. In a most preferred embodiment the level of transcription is determined by techniques selected from the group of Northern blot analysis, reverse transcriptase PCR, real-time PCR, RNAse protection, and microarray. In another embodiment of the invention the kit further comprises means for obtaining a biological sample of the patient. Preferred is a kit, which further comprises a container suitable for containing the means for measuring the level of transcription and the biological sample of the patient, and most preferably further comprises instructions for use and interpretation of the kit results.


In a preferred embodiment the kit for use in classifying biological samples comprises (a) a plurality of oligomers or polynucleotides able to hybridize under stringent or moderately stringent conditions to the transcription products of a gene or genomic region according to Table 1; (b) a container suitable for containing the oligomers or polynucleotides and a biological sample of the patient comprising the transcription products wherein the oligomers or polynucleotide can hybridize under stringent or moderately stringent conditions to the transcription products, (c) means to detect the hybridization of (b); and optionally, (d) instructions for use and interpretation of the kit results.


The kit may also contain other components such as hybridization buffer (where the oligomers are to be used as a probe) packaged in a separate container. Alternatively, where the oligomers are to be used to amplify a target region, the kit may contain, packaged in separate containers, a polymerase and a reaction buffer optimized for primer extension mediated by the polymerase, such as PCR.


Most preferably a kit according to the embodiments of the present invention is used for the determination of expression step of the methods according to other aspects of the invention.


In a particularly preferred embodiment the expression level of the genes, genomic sequences and/or regulatory regions according to Table 1 is determined by analysis of the level of methylation of said genes, genomic sequences and/or regulatory regions thereof. It is preferred that the level of methylation of said genes, genomic sequences and/or regulatory regions thereof is determined by determining the methylation status or level of at least one CpG dinucleotide thereof. It is further preferred that the level of methylation of said genes, genomic sequences and/or regulatory regions thereof is determined by determining the methylation status or level of a plurality of CpG dinucleotides thereof. It is further preferred that the methylation state of CpG positions within regions of the sequences of Table 1, as shown in Table 3 are analyzed. Said analysis comprises the following steps:


a) contacting genomic DNA obtained from the subject with at least one reagent, or series of reagents that distinguishes between methylated and non-methylated CpG dinucleotides within at least one target region of the genomic DNA, wherein said contiguous nucleotides comprise at least one CpG dinucleotide sequence; and


b) classifying the biological sample according to the methylation status of said target regions analyzed in i).


Genomic DNA may be isolated by any means standard in the art, including the use of commercially available kits. Briefly, wherein the DNA of interest is encapsulated in by a cellular membrane the biological sample must be disrupted and lyzed by enzymatic, chemical or mechanical means. The DNA solution may then be cleared of proteins and other contaminants e.g. by digestion with proteinase K. The genomic DNA is then recovered from the solution. This may be carried out by means of a variety of methods including salting out, organic extraction or binding of the DNA to a solid phase support. The choice of method will be affected by several factors including time, expense and required quantity of DNA. Preferably, the source of the DNA sample is selected from the group consisting of cells or cell lines, histological slides, biopsies, paraffin-embedded tissue, bodily fluids, ejaculate, urine, blood, and combinations thereof. Preferably, the source is biopsies, body fluids, ejaculate, urine, or blood. The genomic DNA sample is then treated in such a manner that cytosine bases which are unmethylated at the 5′-position are converted to uracil, thymine, or another base which is dissimilar to cytosine in terms of hybridization behavior. This will be understood as ‘treatment’ herein.


The above described treatment of genomic DNA is preferably carried out with bisulfite (hydrogen sulfite, disulfite) and subsequent alkaline hydrolysis which results in a conversion of non-methylated cytosine nucleobases to uracil or to another base which is dissimilar to cytosine in terms of base pairing behavior.


The treated DNA is then analyzed in order to determine the methylation state of one or more target gene sequences (prior to the treatment) suitable for the classification of biological samples. It is preferred that the target region comprises, or hybridizes under stringent conditions to at least 16 contiguous nucleotides of the converted sequence of at least one gene or genomic sequence selected from the group consisting the genes and genomic sequences as listed in Table 1, and more preferably to the sub-regions thereof according to Table 3. It is preferred that at least one target region is selected from each of Tables 8A to 8L. It is further preferred that the sequences of said genes as described in the accompanying sequence listing are analyzed. The method of analysis may be selected from those known in the art, including those listed herein. Particularly preferred are MethyLight™, MSP™ and the use of blocking oligonucleotides as will be described herein. It is preferred that any oligonucleotides used in such analysis (including primers, blocking oligonucleotides and detection probes) should be reverse complementary, identical, or hybridize under stringent or highly stringent conditions to an at least 16-base-pair long segment of the base sequences of one or more converted sequences selected from Table 2 and sequences complementary thereto.


Further Improvements

The present invention provides novel uses for the genes, genomic sequences and/or regulatory regions thereof according to Table 1. Additional embodiments (see Table 2) provide modified variants of SEQ ID NO: 413 to SEQ ID NO: 824, as well as oligonucleotides and/or PNA-oligomers for analysis of cytosine methylation patterns within the group consisting SEQ ID NO: 413 to SEQ ID NO: 824.


An objective of the invention comprises analysis of the methylation state of one or more CpG dinucleotides within at least one of the genomic sequences selected from the group consisting of SEQ ID NO: 413 to SEQ ID NO: 824 and sequences complementary thereto.


The disclosed invention provides treated nucleic acids, derived from genomic SEQ ID NO: 413 to SEQ ID NO: 824, wherein the treatment is suitable to convert at least one unmethylated cytosine base of the genomic DNA sequence to uracil or another base that is detectably dissimilar to cytosine in terms of hybridization. The genomic sequences in question may comprise one, or more, consecutive or random methylated CpG positions. Said treatment preferably comprises use of a reagent selected from the group consisting of bisulfite, hydrogen sulfite, disulfite, and combinations thereof. In a preferred embodiment of the invention, the objective comprises analysis of a non-naturally occurring modified nucleic acid comprising a sequence of at least 16 contiguous nucleotide bases in length of a sequence selected from the converted sequences of Table 2. It is particularly preferred that said nucleic acid is a non-naturally occurring modified nucleic acid that is not identical to or complementary to the genomic sequences of Table 1 or other human genomic DNA.


It is further preferred that said sequence comprises at least one CpG, TpA or CpA dinucleotide and sequences complementary thereto. The sequences of SEQ ID NO: 1650 to SEQ ID NO: 4120 provide non-naturally occurring modified versions of the nucleic acid according to SEQ ID NO: 413 TO SEQ ID NO: 824, wherein the modification of each genomic sequence results in the synthesis of a nucleic acid having a sequence that is unique and distinct from said genomic sequence as follows. For each sense strand genomic DNA, e.g., SEQ ID NO:1, four converted versions are disclosed. A first version wherein “C” is converted to “T,” but “CpG” remains “CpG” (i.e., corresponds to case where, for the genomic sequence, all “C” residues of CpG dinucleotide sequences are methylated and are thus not converted); a second version discloses the complement of the disclosed genomic DNA sequence (i.e. antisense strand), wherein “C” is converted to “T,” but “CpG” remains “CpG” (i.e., corresponds to case where, for all “C” residues of CpG dinucleotide sequences are methylated and are thus not converted). The ‘upmethylated’ converted sequences of SEQ ID NO: 413 TO SEQ ID NO: 824 correspond to SEQ ID NO: 1650 TO SEQ ID NO: 2472. A third chemically converted version of each genomic sequences is provided, wherein “C” is converted to “T” for all “C” residues, including those of “CpG” dinucleotide sequences (i.e., corresponds to case where, for the genomic sequences, all “C” residues of CpG dinucleotide sequences are unmethylated); a final chemically converted version of each sequence, discloses the complement of the disclosed genomic DNA sequence (i.e. antisense strand), wherein “C” is converted to “T” for all “C” residues, including those of “CpG” dinucleotide sequences (i.e., corresponds to case where, for the complement (antisense strand) of each genomic sequence, all “C” residues of CpG dinucleotide sequences are unmethylated). The ‘downmethylated’ converted sequences of SEQ ID NO: 413 to SEQ ID NO: 824 correspond to SEQ ID NO: 3297 to SEQ ID NO: 4120.


It is particularly preferred that, fragments of the converted DNA are amplified, using sets of primer oligonucleotides according to the present invention, and an amplification enzyme. The amplification of several DNA segments can be carried out simultaneously in one and the same reaction vessel. Typically, the amplification is carried out using a polymerase chain reaction (PCR). The set of primer oligonucleotides includes at least two oligonucleotides whose sequences are each reverse complementary, identical, or hybridize under stringent or highly stringent conditions to an at least 16-base-pair long segment of the base sequences of one of SEQ ID NO: 1650 to SEQ ID NO: 4120 and sequences complementary thereto.


In an alternate embodiment of the method, the methylation status of preselected CpG positions within the nucleic acid sequences comprising one or more of SEQ ID NO: 413 to SEQ ID NO: 824 may be detected by use of methylation-specific primer oligonucleotides. This technique (MSP) has been described in U.S. Pat. No. 6,265,171 to Herman. The use of methylation status specific primers for the amplification of bisulfite converted DNA allows the differentiation between methylated and unmethylated nucleic acids. MSP primers pairs contain at least one primer which hybridizes to a bisulfite converted CpG dinucleotide. Therefore, the sequence of said primers comprises at least one CpG dinucleotide. MSP primers specific for non-methylated DNA contain a “T’ at the position of the C position in the CpG. Preferably, therefore, the base sequence of said primers is required to comprise a sequence having a length of at least 9 nucleotides which hybridizes to a converted nucleic acid sequence according to one of SEQ ID NO: 1650 to SEQ ID NO: 4120 and sequences complementary thereto, wherein the base sequence of said oligomers comprises at least one CpG dinucleotide.


A further preferred embodiment of the method comprises the use of blocker oligonucleotides. The use of such blocker oligonucleotides has been described by Yu et al., BioTechniques 23:714-720, 1997. Blocking probe oligonucleotides are hybridized to the bisulfite converted nucleic acid concurrently with the PCR primers. PCR amplification of the nucleic acid is terminated at the 5′ position of the blocking probe, such that amplification of a nucleic acid is suppressed where the complementary sequence to the blocking probe is present. The probes may be designed to hybridize to the bisulfite converted nucleic acid in a methylation status specific manner. For example, for detection of methylated nucleic acids within a population of unmethylated nucleic acids, suppression of the amplification of nucleic acids which are unmethylated at the position in question would be carried out by the use of blocking probes comprising a ‘CpA’ or ‘TpA’ at the position in question, as opposed to a ‘CpG’ if the suppression of amplification of methylated nucleic acids is desired.


For PCR methods using blocker oligonucleotides, efficient disruption of polymerase-mediated amplification requires that blocker oligonucleotides not be elongated by the polymerase. Preferably, this is achieved through the use of blockers that are 3′-deoxyoligonucleotides, or oligonucleotides derivitized at the 3′ position with other than a “free” hydroxyl group. For example, 3′-O-acetyl oligonucleotides are representative of a preferred class of blocker molecule.


Additionally, polymerase-mediated decomposition of the blocker oligonucleotides should be precluded. Preferably, such preclusion comprises either use of a polymerase lacking 5′-3′ exonuclease activity, or use of modified blocker oligonucleotides having, for example, thioate bridges at the 5′-terminii thereof that render the blocker molecule nuclease-resistant. Particular applications may not require such 5′ modifications of the blocker. For example, if the blocker- and primer-binding sites overlap, thereby precluding binding of the primer (e.g., with excess blocker), degradation of the blocker oligonucleotide will be substantially precluded. This is because the polymerase will not extend the primer toward, and through (in the 5′-3′ direction) the blocker—a process that normally results in degradation of the hybridized blocker oligonucleotide.


A particularly preferred blocker/PCR embodiment, for purposes of the present invention and as implemented herein, comprises the use of peptide nucleic acid (PNA) oligomers as blocking oligonucleotides. Such PNA blocker oligomers are ideally suited, because they are neither decomposed nor extended by the polymerase.


Preferably, therefore, the base sequence of said blocking oligonucleotides is required to comprise a sequence having a length of at least 9 nucleotides which hybridizes to a converted nucleic acid sequence according to one of SEQ ID NO: 1650 to SEQ ID NO: 4120 and sequences complementary thereto, wherein the base sequence of said oligonucleotides comprises at least one CpG, TpG or CpA dinucleotide.


The fragments obtained by means of the amplification can carry a directly or indirectly detectable label. Preferred are labels in the from of fluorescence labels, radionuclides, or detachable molecule fragments having a typical mass which can be detected in a mass spectrometer. Where said labels are mass labels, it is preferred that the labeled amplificates have a single positive or negative net charge, allowing for better detectability in the mass spectrometer. The detection may be carried out and visualized by means of, e.g., matrix assisted laser desorption/ionization mass spectrometry (MALDI) or using electron spray mass spectrometry (ESI).


Matrix Assisted Laser Desorption/Ionization Mass Spectrometry (MALDI-TOF) is a very efficient development for the analysis of biomolecules (Karas and Hillenkamp, Anal Chem., 60:2299-301, 1988). An analyte is embedded in a light-absorbing matrix. The matrix is evaporated by a short laser pulse thus transporting the analyte molecule into the vapour phase in an unfragmented manner. The analyte is ionized by collisions with matrix molecules. An applied voltage accelerates the ions into a field-free flight tube. Due to their different masses, the ions are accelerated at different rates. Smaller ions reach the detector sooner than bigger ones. MALDI-TOF spectrometry is well suited to the analysis of peptides and proteins. The analysis of nucleic acids is somewhat more difficult (Gut and Beck, Current Innovations and Future Trends, 1:147-57, 1995). The sensitivity with respect to nucleic acid analysis is approximately 100-times less than for peptides, and decreases disproportionately with increasing fragment size. Moreover, for nucleic acids having a multiply negatively charged backbone, the ionization process via the matrix is considerably less efficient. In MALDI-TOF spectrometry, the selection of the matrix plays an eminently important role. For desorption of peptides, several very efficient matrixes have been found which produce a very fine crystallisation. There are now several responsive matrixes for DNA, however, the difference in sensitivity between peptides and nucleic acids has not been reduced. This difference in sensitivity can be reduced, however, by chemically modifying the DNA in such a manner that it becomes more similar to a peptide. For example, phosphorothioate nucleic acids, in which the usual phosphates of the backbone are substituted with thiophosphates, can be converted into a charge-neutral DNA using simple alkylation chemistry (Gut and Beck, Nucleic Acids Res. 23: 1367-73, 1995). The coupling of a charge tag to this modified DNA results in an increase in MALDI-TOF sensitivity to the same level as that found for peptides. A further advantage of charge tagging is the increased stability of the analysis against impurities, which makes the detection of unmodified substrates considerably more difficult.


In the next step of the method, the amplificates obtained are analyzed in order to ascertain the methylation status of the CpG dinucleotides prior to the treatment.


In embodiments where the amplificates were obtained by means of MSP amplification, the presence or absence of an amplificate is in itself indicative of the methylation state of the CpG positions covered by the primer, according to the base sequences of said primer.


Amplificates obtained by means of both standard and methylation specific PCR may be further analyzed by means of hybridization-based methods such as, but not limited to, array technology and probe based technologies as well as by means of techniques such as sequencing and template directed extension.


In one embodiment of the method, the amplificates synthesised are subsequently hybridized to an array or a set of oligonucleotides and/or PNA oligomers. In this context, the hybridization takes place in the following manner: the set of probes used during the hybridization is preferably composed of at least 2 oligonucleotides or PNA-oligomers; in the process, the amplificates serve as probes which hybridize to oligonucleotides or PNA oligomers previously bonded to a solid phase; the non-hybridized fragments are subsequently removed; said oligonucleotides or PNA oligomers contain at least one base sequence having a length of at least 9 nucleotides which is reverse complementary or identical to a segment of a sequence selected from SEQ ID NO: 1650 to SEQ ID NO: 4120; and the segment comprises at least one CpG, TpG or CpA dinucleotide.


In a preferred embodiment, said dinucleotide is present in the central third of the oligomer. For example, wherein the oligomer comprises one CpG dinucleotide, said dinucleotide is preferably the fifth to ninth nucleotide from the 5′-end of a 13-mer. One oligonucleotide exists for the analysis of each CpG dinucleotide within the sequence according to SEQ ID NO: 413 TO SEQ ID NO: 4120. It is preferred that at least one oligonucleotide is used to determine the status of at least one CpG dinucleotide of a gene selected from each of Tables 8A to 8L. Said oligonucleotides may also be present in the form of peptide nucleic acids. The non-hybridized amplificates are then removed. The hybridized amplificates are then detected. In this context, it is preferred that labels attached to the amplificates are identifiable at each position of the solid phase at which an oligonucleotide sequence is located.


In a particularly preferred embodiment of the method, the genomic methylation status of the CpG positions may be ascertained by means of oligonucleotide probes that are hybridized to the bisulfite converted DNA concurrently with the PCR amplification primers (wherein said primers may either be methylation specific or standard).


A particularly preferred embodiment of this method is the use of fluorescence-based Real Time Quantitative PCR (Heid et al., Genome Res. 6:986-994, 1996; also see U.S. Pat. No. 6,331,393) employing a dual-labeled fluorescent oligonucleotide probe (TaqMan™ PCR, using an ABI Prism 7700 Sequence Detection System, Perkin Elmer Applied Biosystems, Foster City, Calif.). The TaqMan™ PCR reaction employs the use of a nonextendible interrogating oligonucleotide, called a TaqMan™ probe, which, in preferred embodiments, is designed to hybridize to a GpC-rich sequence located between the forward and reverse amplification primers. The TaqMan™ probe further comprises a fluorescent “reporter moiety” and a “quencher moiety” covalently bound to linker moieties (e.g., phosphoramidites) attached to the nucleotides of the TaqMan™ oligonucleotide. For analysis of methylation within nucleic acids subsequent to bisulfite treatment, it is required that the probe be methylation specific, as described in U.S. Pat. No. 6,331,393, (hereby incorporated by reference in its entirety) also known as the MethyLight™ assay. Variations on the TaqMan™ detection methodology that are also suitable for use with the described invention include the use of dual-probe technology (Lightcycler™) or fluorescent amplification primers (Sunrise™ technology). Both these techniques may be adapted in a manner suitable for use with bisulfite converted DNA, and moreover for methylation analysis within CpG dinucleotides.


A further suitable method for the use of probe oligonucleotides for the assessment of methylation by analysis of bisulfite converted nucleic acids In a further preferred, the method comprises the use of template-directed oligonucleotide extension, such as MS-SNuPE as described by Gonzalgo and Jones, Nucleic Acids Res. 25:2529-2531, 1997.


In yet a further embodiment of the method, said step comprises sequencing and subsequent sequence analysis of the amplificate (Sanger F., et al., Proc Natl Acad Sci USA 74:5463-5467, 1977).


In a preferred embodiment, the methylation analysis comprises the use of an oligonucleotide or oligomer for detecting the cytosine methylation state within genomic or treated (chemically modified) DNA, according to SEQ ID NO: 413 to SEQ ID NO: 4120. Said oligonucleotide or oligomer comprising a nucleic acid sequence having a length of at least nine (9) nucleotides which hybridizes, under moderately stringent or stringent conditions (as defined herein above), to a treated nucleic acid sequence according to SEQ ID NO: 1650 to SEQ ID NO: 4120 and/or sequences complementary thereto, or to a genomic sequence according to SEQ ID NO: 413 to SEQ ID NO: 824 and/or sequences complementary thereto.


Thus, the present invention includes nucleic acid molecules (e.g., oligonucleotides and peptide nucleic acid (PNA) molecules (PNA-oligomers)) that hybridize under moderately stringent and/or stringent hybridization conditions to all or a portion of the sequences SEQ ID NO: 413 to SEQ ID NO: 4120, or to the complements thereof. Particularly preferred is a nucleic acid molecule that hybridizes under moderately stringent and/or stringent hybridization conditions to all or a portion of the sequences SEQ ID NO: 1650 to SEQ ID NO: 4120 but is not identical to or complementary to the equivalent genomic DNA selected from SEQ ID NO: 413 to SEQ ID NO: 4120 or other human genomic DNA. The hybridizing portion of the hybridizing nucleic acids is typically at least 9, 15, 20, 25, 30 or 35 nucleotides in length. However, longer molecules have inventive utility, and are thus within the scope of the present invention.


Preferably, the hybridizing portion of the inventive hybridizing nucleic acids is at least 95%, or at least 98%, or 100% identical to the sequence, or to a portion thereof of SEQ ID NO: 413 to SEQ ID NO: 4120, or to the complements thereof.


Hybridizing nucleic acids of the type described herein can be used, for example, as a primer (e.g., a PCR primer), or a probe. Preferably, hybridization of the oligonucleotide probe to a nucleic acid sample is performed under stringent conditions and the probe is 100% identical to the target sequence.


Nucleic acid duplex or hybrid stability is expressed as the melting temperature or Tm, which is the temperature at which a probe dissociates from a target DNA. This melting temperature is used to define the required stringency conditions.


For target sequences that are related and substantially identical to the corresponding sequence of SEQ ID NO: 413 to SEQ ID NO: 4120 (such as allelic variants and SNPs), rather than identical, it is useful to first establish the lowest temperature at which only homologous hybridization occurs with a particular concentration of salt (e.g., SSC or SSPE). Then, assuming that 1% mismatching results in a 1° C. decrease in the Tm, the temperature of the final wash in the hybridization reaction is reduced accordingly (for example, if sequences having >95% identity with the probe are sought, the final wash temperature is decreased by 5° C.). In practice, the change in Tm can be between 0.5° C. and 1.5° C. per 1% mismatch.


Examples of inventive oligonucleotides of length X (in nucleotides), as indicated by polynucleotide positions with reference to, e.g., SEQ ID NO:1, include those corresponding to sets (sense and antisense sets) of consecutively overlapping oligonucleotides of length X, where the oligonucleotides within each consecutively overlapping set (corresponding to a given X value) are defined as the finite set of Z oligonucleotides from nucleotide positions:


n to (n+(X−1));


where n=1, 2, 3, . . . (Y−(X−1));


where Y equals the length (nucleotides or base pairs) of SEQ ID NO: 1 (444);


where X equals the common length (in nucleotides) of each oligonucleotide in the set (e.g., X=20 for a set of consecutively overlapping 20-mers); and


where the number (Z) of consecutively overlapping oligomers of length X for a given SEQ ID NO of length Y is equal to Y−(X−1). For example Z=444−19=425 for either sense or antisense sets of SEQ ID NO:1, where X=20.


Preferably, the set is limited to those oligomers that comprise at least one CpG, TpG or CpA dinucleotide.


Examples of inventive 20-mer oligonucleotides include the following set of oligomers (and the antisense set complementary thereto), indicated by polynucleotide positions with reference to SEQ ID NO:1:

    • 1-20, 2-21, 3-22, 4-23, 5-24, . . . 425-444.


Preferably, the set is limited to those oligomers that comprise at least one CpG, TpG or CpA dinucleotide.


Likewise, examples of inventive 25-mer oligonucleotides include the following set of oligomers (and the antisense set complementary thereto), indicated by polynucleotide positions with reference to SEQ ID NO:1:

    • 1-25, 2-26, 3-27, 4-28, 5-29, . . . 420-444.


Preferably, the set is limited to those oligomers that comprise at least one CpG, TpG or CpA dinucleotide.


The present invention encompasses, for each of SEQ ID NO: 413 TO SEQ ID NO: 4120 (sense and antisense), multiple consecutively overlapping sets of oligonucleotides or modified oligonucleotides of length X, where, e.g., X=9, 10, 17, 20, 22, 23, 25, 27, 30 or 35 nucleotides.


The oligonucleotides or oligomers according to the present invention constitute effective tools useful to ascertain genetic and epigenetic parameters of the genomic sequence corresponding to SEQ ID NO: 413 to SEQ ID NO: 824. Preferably, said oligomers comprise at least one CpG, TpG or CpA dinucleotide.


Particularly preferred oligonucleotides or oligomers according to the present invention are those in which the cytosine of the CpG dinucleotide (or of the corresponding converted TpG or CpA dinculeotide) sequences is within the middle third of the oligonucleotide; that is, where the oligonucleotide is, for example, 13 bases in length, the CpG, TpG or CpA dinucleotide is positioned within the fifth to ninth nucleotide from the 5′-end.


The oligonucleotides of the invention can also be modified by chemically linking the oligonucleotide to one or more moieties or conjugates to enhance the activity, stability or detection of the oligonucleotide. Such moieties or conjugates include chromophores, fluorophors, lipids such as cholesterol, cholic acid, thioether, aliphatic chains, phospholipids, polyamines, polyethylene glycol (PEG), palmityl moieties, and others as disclosed in, for example, U.S. Pat. Nos. 5,514,758, 5,565,552, 5,567,810, 5,574,142, 5,585,481, 5,587,371, 5,597,696 and 5,958,773. The probes may also exist in the from of a PNA (peptide nucleic acid) which has particularly preferred pairing properties. Thus, the oligonucleotide may include other appended groups such as peptides, and may include hybridization-triggered cleavage agents (Krol et al., BioTechniques 6:958-976, 1988) or intercalating agents (Zon, Pharm. Res. 5:539-549, 1988). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a chromophore, fluorophor, peptide, hybridization-triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc.


The oligonucleotide may also comprise at least one art-recognized modified sugar and/or base moiety, or may comprise a modified backbone or non-natural internucleoside linkage.


The oligonucleotides or oligomers according to particular embodiments of the present invention are typically used in ‘sets,’ which contain at least one oligomer for analysis of at least one of the CpG dinucleotides of genomic sequences SEQ ID NO: 413 to SEQ ID NO: 824 and sequences complementary thereto, or to the corresponding CpG, TpG or CpA dinucleotide within a sequence of the treated nucleic acids according to SEQ ID NO: 1650 to SEQ ID NO: 4120 and sequences complementary thereto. However, it is anticipated that for economic or other factors it may be preferable to analyze a limited selection of the CpG dinucleotides within said sequences, and the content of the set of oligonucleotides is altered accordingly.


Therefore, in particular embodiments, the present invention provides a set of at least two (2) (oligonucleotides and/or PNA-oligomers) useful for detecting the cytosine methylation state in treated genomic DNA (SEQ ID NO: 1650 to SEQ ID NO: 4120), or in genomic DNA (SEQ ID NO: 413 to SEQ ID NO: 824) and sequences complementary thereto. These probes enable the classification of biological samples. The set of oligomers may also be used for detecting single nucleotide polymorphisms (SNPs) in treated genomic DNA (SEQ ID NO: 1650 to SEQ ID NO: 4120), or in genomic DNA (SEQ ID NO: 413 to SEQ ID NO: 824 and sequences complementary thereto).


In preferred embodiments, at least one, and more preferably all members of a set of oligonucleotides is bound to a solid phase.


In further embodiments, the present invention provides a set of at least two (2) oligonucleotides that are used as ‘primer’ oligonucleotides for amplifying DNA sequences of one of SEQ ID NO: 413 to SEQ ID NO: 4120 and sequences complementary thereto, or segments thereof.


It is anticipated that the oligonucleotides may constitute all or part of an “array” or “DNA chip” (i.e., an arrangement of different oligonucleotides and/or PNA-oligomers bound to a solid phase). Such an array of different oligonucleotide- and/or PNA-oligomer sequences can be characterized, for example, in that it is arranged on the solid phase in the from of a rectangular or hexagonal lattice. The solid-phase surface may be composed of silicon, glass, polystyrene, aluminum, steel, iron, copper, nickel, silver, or gold. Nitrocellulose as well as plastics such as nylon, which can exist in the from of pellets or also as resin matrices, may also be used. An overview of the Prior Art in oligomer array manufacturing can be gathered from a special edition of Nature Genetics (Nature Genetics Supplement, Volume 21, January 1999, and from the literature cited therein). Fluorescently labeled probes are often used for the scanning of immobilized DNA arrays. The simple attachment of Cy3 and Cy5 dyes to the 5′-OH of the specific probe are particularly suitable for fluorescence labels. The detection of the fluorescence of the hybridized probes may be carried out, for example, via a confocal microscope. Cy3 and Cy5 dyes, besides many others, are commercially available.


It is also anticipated that the oligonucleotides, or particular sequences thereof, may constitute all or part of an “virtual array” wherein the oligonucleotides, or particular sequences thereof, are used, for example, as ‘specifiers’ as part of, or in combination with a diverse population of unique labeled probes to analyze a complex mixture of analytes. Such a method, for example is described in US 2003/0013091 (U.S. Ser. No. 09/898,743, published 16 Jan. 2003). In such methods, enough labels are generated so that each nucleic acid in the complex mixture (i.e., each analyte) can be uniquely bound by a unique label and thus detected (each label is directly counted, resulting in a digital read-out of each molecular species in the mixture).


It is particularly preferred that the oligomers according to the invention are utilised for at least one of: in determining the presence or absence of specific organ, tissue or cell types, the detection and/or classification of a cell proliferative disorder and/or analysis of cellular differentiation.


In one embodiment of the method, this is achieved by analysis of the methylation status of at least one target sequence comprising, or hybridizing under stringent conditions to at least 16 contiguous nucleotides of a gene or sequence selected from the group consisting the genes and sequences according to Table 1 and complements thereof.


In a particularly preferred embodiment of the invention, the expression at least one of the genes, genomic sequences and/or regulatory regions thereof from each of Tables 8A to 8L is used in the classification of said sample. It is particularly preferred that said biological sample is classified according to at least one parameter selected from the group consisting of the cell, organ or tissue type of said biological sample or features thereof such as disease state.


Accordingly, in a particularly preferred embodiment the invention provides a method for the classification of biological samples, comprising the following steps:


a) determining the expression levels of one or more genes or gene sequences of each of Tables 8A to 8L and/or regulatory regions thereof; and


b) classifying said biological sample according to said expression status.


In a the most preferred embodiment thereof, the present invention provides a method for classifying a biological sample, comprising: obtaining a biological sample comprising genomic nucleic acid(s); contacting the nucleic acid(s), or a fragment thereof, with one reagent or a plurality of reagents sufficient for distinguishing between methylated and non methylated CpG dinucleotide sequences within a target sequence of the subject nucleic acid, wherein the target sequence comprises, or hybridizes under stringent conditions to, a sequence comprising at least 16 contiguous nucleotides of at least one gene or genomic sequence selected from each of Tables 8A to 8L said contiguous nucleotides comprising at least one CpG dinucleotide sequence; and determining, based at least in part on said distinguishing, the methylation state of at least one target CpG dinucleotide sequence, or an average, or a value reflecting an average methylation state of a plurality of target CpG dinucleotide sequences.


Preferably, distinguishing between methylated and non methylated CpG dinucleotide sequences within the target sequence comprises methylation state-dependent conversion or non-conversion of at least one such CpG dinucleotide sequence to the corresponding converted or non-converted dinucleotide sequence within a sequence selected from the equivalent converted sequence selected from Tables 8A to 8L, and contiguous regions thereof corresponding to the target sequence.


In the final step of the method the classification of the biological sample according to the measured expression is determined. Table 6 and 7 provide information for the correlation of measured methylation with cell, tissue and/or organ types. The person skilled in the art will be able to interpret RNA and/or protein expression on this basis. It is generally appreciated that there is an inverse correlation between methylation and mRNA and/or polypeptide expression.


A person with ordinary skills in the art will be able to utilize the information provided in Tables 6 to 9 in order to select suitable markers according to his specific requirements. The information provided in said tables enables the selection of one or a plurality of genes or genomic sequences in order to enable the classification of biological samples.


The information provided in Tables 6 to 9 will also enable a person with ordinary skills in the art to combine suitable markers in order to classify or identify of biological samples. This can be done for example, but not limited to, by combing at least two genes or genomics regions which have a complementary and/or overlapping expression pattern. In one embodiment of the invention the information provided by Table 6 and 7 is used to for the classification of one or more biological samples. In a particularly preferred embodiment the information of Table 6 is used for the classification of a biological sample according to cell, tissue and/or organ type. In an alternative embodiment the information according to Table 7 is used for the conformation and/or monitoring of cell, tissue and/or organ type which were derived by means of cell culturing or tissue engineering processes.


Kits

The described invention further provides a composition of matter useful for the classification of biological samples. Said composition comprising at least one nucleic acid 18 base pairs in length of a segment of a nucleic acid sequence selected from the group consisting SEQ ID NO: 1650 TO SEQ ID NO: 4120, and one or more substances taken from the group comprising: magnesium chloride, dNTP, Taq polymerase, bovine serum albumen, an oligomer in particular an oligonucleotide or peptide nucleic acid (PNA)-oligomer, said oligomer comprising in each case at least one base sequence having a length of at least 9 nucleotides which is complementary to, or hybridizes under moderately stringent or stringent conditions to a pretreated genomic DNA according to one of SEQ ID NO: 1650 TO SEQ ID NO: 4120 and sequences complementary thereto. It is preferred that said composition of matter comprises a buffer solution appropriate for the stabilization of said nucleic acid in an aqueous solution and enabling polymerase based reactions within said solution. Suitable buffers are known in the art and commercially available.


Utility

The subject matter of the invention has specific utility in the fields of medicine and/or molecular biology. In particular aspects, the subject matter are one or more markers or comprises at least parts of one or more markers. Thereby a marker is a gene, a genomic sequence, a regulatory region of a gene according to Table 1 or its mRNA, cDNA or polypeptide (protein, peptide). herein also referred as molecular biological marker. According to the invention, the provided markers have novel utility for the classification of biological samples.


A utility of the present invention is to provide molecular markers and methods for the analysis thereof that may be considered an alternative to traditional histological or pathological analysis. Said molecular biological markers accordingly offer an alternative to current means such as staining and microscopic analysis.


The present invention, in particular said markers are of use in determining the presence or absence of specific organ, tissue or cell types in a biological sample. Wherein said sample is heterogeneous in nature, the method according to the present invention may be used for the identification of a population or subpopulation of specific organs, tissue or cell types. One application of this is for the determination of the tumor content of a biopsy sample which may heterogeneously comprise both tumor and normal tissue. The determination of the relative tumor content of sample is of particular interest when quantifying the presence of molecular markers by reference to the tumor content of the sample, as for example described in EP 05090318 (which is hereby incorporated by reference in its entirety).


Another application of the determination of the presence or absence of specific organ, tissue or cell types in a biological sample by means of the present invention, in particular said markers is the identification of the tissue origin of tumors or metastasis of unknown tissue origin (cancer of unknown primary, CUP). The treatment of tumors of unknown identity is a well known problem. 3-5% of all cancer diseases are cancer of unknown primary. The course of disease is characterized in that only metastasis of unknown primary tumor are detected. A tumor-specific treatment is therefore not possible. The prognosis for the affected individual is bad and the survival rate is correspondingly low. So far mainly expression-based methods for the identification of a tumor's origin are known. These methods are highly error-associated and difficult because RNA degrades quickly and easily. Therefore an exact determination of the RNA is often not possible. In addition, also a correspondent reference has to be included. But according to the present invention, the origin of tumor cells is easily determinable. Thereby genomic DNA is isolated from a fresh sample or from an archived sample. For example but not limited to, the archived sample is a formalin-fixed and/or paraffin-embedded sample. The isolated DNA is then subjected to a bisulfite conversion. Suitable methods for DNA isolation and bisulfite conversion are known in the art, for example, but not limited to, see WO 06/039563. Subsequently the converted DNA is subjected to a real time PCR based detection assays specific for the said markers. Suitable methods for real time PCR based assays are known in the art, for example but not limited to it, such an assay is a QM assay, HM assay or a MSP assay. The origin of the tumor is the identified when a tissue marker is identified which does not belong to the tissue from whom the sample was taken. Alternatively, the isolated DNA is subjected to non-real time PCR amplifications which are specific for said markers. The resulting amplicons are then detected. Suitable method for detection are known in the art. For example but not limited to it, gel electrophoresis, fluorescence, or detection by means of hybridization, for example but not limited to it, to an array. In addition, it is also possible to detect the origin of a tumor only by analyzing the genomic DNA derived from blood. Because tumors are characterized by a high rate of cell death, in particular after chemotherapy, the amount of tumor derived genomic DNA within the blood is increased in comparison to normal levels of tissue derived DNA. Therefore it is sufficient to determine if the DNA specific for a tissue is increased in the blood of a patient with cancer of unknown primary. The tissue which shows an increased level of DNA in the blood is the tissue of origin of the tumor. For this application, the said markers of the invention are used essentially according to WO 03/074730. According to it, a body fluid sample is obtained from an individual, the amount or presence of free floating DNA originating from a tissue or organ is determinated, and the presence or absence of a medical condition is determinated based on the amount or presence of the free floating DNA originating from a tissue or organ. In any case, after identification of the tumors or metastasis origin, the treatment can be adjusted. The identification also enables that the tumor can be identified in its primary tissue, from where it can then be removed. The use of the present invention, in particular said markers has several advantages in comparison to the methods for tissue identification based on expression, in particular RNA based detection: (a) DNA and DNA methylation are much more stable then RNA; (b) DNA is upstream of the regulatory cascade. This means that DNA methylation influences a lot of RNA expression. Therefore usually the analysis of a complete panel of different RNAs is necessary, while only one DNA methylation pattern in many times sufficient.


The present invention, in particular said markers have further utility in the detection and/or classification of a cell proliferative disorder, for example but not limited to cancer. It is known in the art that increased levels of circulating cells or cellular matter are a characteristic of cell proliferative disorders. The methods or markers according to the present invention enable the detection and identification of atypical levels of cells or cellular matter derived from specific organ, tissue or cell types and thereby enable the determination of the primary location of said proliferative disorder. They also enable the detection and identification of atypical levels of expression which is a sign of dedifferentiation and breakdown of the cellular regulation mechanisms. Furthermore wherein the presence of a proliferative disorder has already been detected the primary location thereof may be determined according to the methods of the present invention.


The methods or markers of the present invention have a further alternative utility in the analysis of cellular differentiation, for example in the field of tissue engineering. The molecular characterization of a biological sample as opposed to traditional histological analysis enables the improved monitoring of differentiating cell or tissue cultures. The invention solves this longstanding need in the art by providing markers i.e. genes, genomic sequences and/or regulatory regions thereof according to Table 1 (or to one or more of those), the expression thereof at the mRNA, cDNA, protein or peptide level being indicative of the class of said biological sample.


In particular, the genes, genomic sequences and/or regulatory regions thereof according to Table 8A are of use in the differentiation and/or detection of T-lymphocytes. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8B are of use in the differentiation and/or detection of embryonic liver. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8C are of use in the differentiation and/or detection of embryonic skeletal muscle. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8D are of use in the differentiation and/or detection of fibroblasts. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8E are of use in the differentiation and/or detection of heart muscle. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8F are of use in the differentiation of heart muscle from skeletal muscle. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8G are of use in the differentiation and/or detection of keratinocytes. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8H are of use in the differentiation and/or detection of liver. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8I are of use in the differentiation and/or detection of melanocytes. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8J are of use in the differentiation and/or detection of placenta. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8K are of use in the differentiation and/or detection of skeletal muscle. In particular the genes, genomic sequences and/or regulatory regions thereof according to Table 8L are of use in the differentiation and/or detection of sperm.


The genomic sequences (and corresponding genes) according to Table 9 are of particular use in the fields of cell culture and tissue engineering, for the confirmation and/or monitoring of particular cell, tissue and/organ types. In particular the genomic sequences according to Table 9A are suitable for the confirmation and/or monitoring of T-lymphocytes. In particular the genomic sequences according to Table 9B are suitable for the confirmation and/or monitoring of embryonic liver. In particular the genomic sequences according to Table 9C are suitable for the confirmation and/or monitoring of embryonic skeletal muscle. In particular the genomic sequences according to Table 9D are suitable for the confirmation and/or monitoring of fibroblasts. In particular the genomic sequences according to Table 9E are suitable for confirmation and/or monitoring of heart muscle. In particular the genomic sequences according to Table 9F are suitable for the confirmation and/or monitoring of keratinocytes. In particular the genomic sequences according to Table 9G are suitable for the confirmation and/or monitoring of liver. In particular the genomic sequences according to Table 9H are suitable for the confirmation and/or monitoring of melanocytes. In particular the genomic sequences according to Table 9I are suitable for the confirmation and/or monitoring of placenta. In particular the genomic sequences according to Table 9J are suitable for the confirmation and/or monitoring of skeletal muscle.


According to a particularly preferred embodiment of the invention, the methylation status of CpG positions of genes, genomic sequences and/or regulatory regions thereof according to Table 1 is used in the classification of said sample. It is particularly preferred that said biological sample is classified according to at least one parameter selected from the group consisting of the cell, organ or tissue type of said biological sample or features thereof such as disease state. According to a particular preferred embodiment, the subject matter of the invention, in particular the methods, compositions, kits, or markers are utilised for at least one of histological analysis, pathological analysis, detection and/or characterization of cell proliferative disorders and monitoring of cellular or tissue differentiation.


According to the invention, the provided markers, in particular the genes, genomic sequences, regulatory regions, and corresponding mRNAs, cDNAs, proteins or peptides have a particular utility in the following aspects. Thereby a single marker is used either alone or in combination with other marker or markers herein provided or not.


The herein provided markers have utility (i) for the characterization of the marker corresponding tissue or cell, (ii) for the identification of marker corresponding tissue or cell, (iii) for the isolation of marker corresponding tissue or cell, (iv) for the purification of the corresponding tissue or cell, or (v) combinations thereof. Therefore known methods, so far unreported methods, or combinations thereof are useable. Said application is useful in the field of research, diagnostics as well as therapeutics.


In addition, the herein provided markers have utility for the prospective profiling, retrospective profiling, or both of donors and/or recipients in organ transplantation procedures. The correct characterization, identification, or both of the donor and/or the recipient is mandatory during organ transplantation procedures to assure the success of the intervention. The use of the markers of the invention enables the profiling of both, donor and recipient, form which prospective or retrospective observations or conclusions about the feasibility of the procedure are drawn.


In addition, the herein provided markers have utility for histological, chemical and/or immunohistochemical analysis. Accordingly, they have utility in the fields of research as well as diagnostics, in particular for histological or pathological analysis.


In addition, the herein provided markers have utility for phylogenetic profiling of species or tissues. The ontogenetic origin or the developmental lineage is then determined by comparison of the determined profiles.


In addition, the herein provided markers have utility for quality control of a genetically modified organism, tissue, group of cells or cell.


In addition, the herein provided markers have utility for controlling side effects in in vivo gene therapy procedures wherein genetically modified organism, tissue, group of cells or cell is used.


In addition, the herein provided markers have utility for the characterization, identification, or labelling of corresponding tissue or combinations thereof. This is of particular utility in the field of tissue bank storage and proliferation. Furthermore it has utility in a prospective as well as in a retrospective manner. The provided markers allow the individualization of samples by a precise molecular method. This is mandatory in storing biological material from patients or healthy individuals. In addition, this also advantageous for isolation or purification of tissues cells.


In addition, the herein provided markers have utility for controlling cell differentiation in stem-cell research and/or therapeutics. Cells undergo many genetic and/or epigenetic changes throughout differentiation. These changes influence the physiology of the cell and their control is mandatory in any procedure involving stem-cell in research and/or therapeutics. The provided markers allow to control this changes by giving a reference of the adult (completely differentiated) and embryonic (partially differentiated) status of the cells.


CD4+ and CD8+ Lymphocytes:

The herein provided markers of Table 8A and Table 9A have utility for the quantification of lymphocytes, in particular in peripheral blood. The said markers enable the identification of CD4+ and CD8+ lymphocytes among other cells in blood samples. A low number of leucocytes in blood (leucopenia) may indicate bone marrow failure (for example, due to infection, tumor, fibrosis); presence of cytotoxic substance; collagen-vascular diseases (such as lupus erythematosus); disease of the liver or spleen; or radiation. A high number of leucocytes in blood (leucocytosis) may indicate infectious diseases; inflammatory disease (such as rheumatoid arthritis or allergy); leukemia; severe emotional or physical stress; tissue damage (for example, burns); or anemia.


In addition, the herein provided markers of Table 8A and Table 9A have utility for the study of CD4 and/or CD8 T-lymphocyte infiltration in other tissues healthy or diseased. Infiltration of lymphocytes in healthy or diseased tissues is an indication of several diseases such immunological malignances or even in tumor development. The said markers represent a target for the development of molecular probes that coupled to any detection method (e.g. Fluorescent dye) allow the identification of these cells in histological preparations.


In addition, the herein provided markers of Table 8A and Table 9A have utility for identification, isolation and/or purification of CD4 T-lymphocytes and/or CD8 T-lymphocytes, in particular from surrounding tissue infiltrated by the T-lymphocytes; from blood; and/or from other body fluids.


In addition, the herein provided markers of Table 8A and Table 9A have utility for the identification of an individual. Thereby at least two samples are used. One samples is obtained from an individual and another sample is a forensic sample, in particular traces of body cells, tissues or fluids, for example but not limited to, traces of blood and/or body fluids. This is of particular utility in the field of forensic medicine or of legal medicine. As constituent of blood or body fluids, CD4 T-lymphocytes and CD8 T-lymphocytes are part of the mentioned traces. The said markers have the advantage of being stable over time because they are DNA based. In addition said markers have the advantage that they enable a highly detailed and accurate characterization of samples. Through this an unambiguous identification of an individual is enabled.


In addition, the herein provided markers of Table 8A and Table 9A have utility for diagnosing the presence or absence of a disease. Thereby the number of CD4 T-lymphocytes, CD8 T-lymphocytes or both is quantified in normalized samples of healthy individuals. The determined number of CD4 T-lymphocytes, CD8 T-lymphocytes or both are then considered as indicative for healthy condition or a diseased condition with respect to an individual. Preferably, large amount of normalized samples are considered to generate reference values of CD4 T-lymphocytes, CD8 T-lymphocytes or both for a healthy condition and/or for one or more diseased conditions. The diseased condition can be any kind of diseased condition. Preferably, the diseased condition is a disease which causes a immune reaction. For example but not limited to the diseased condition is a cancer disease, a cell proliferation disease, or HIV. Preferably the total number of cells present in a sample is determined. The number of CD4 T-lymphocytes, CD8 T-lymphocytes or both are then normalized to the total number of cells.


Embryonic

The herein provided markers of Table 8B, Table 8C, Table 9B and Table 9C have utility for the study, identification and/or quantification of fetal cells or fetal DNA circulating in maternal blood and/or amniotic fluid. During pregnancy cells and DNA from the fetus are continuously brought to the maternal blood stream as well as the amniotic fluid. Fetal cells and fetal DNA have a diagnostic potential in monitoring the health status of the fetus as reviewed by Bianchi D, 2004 (Bianchi D W. Circulating fetal DNA: its origin and diagnostic potential-a review. Placenta. 2004 April; 25 Suppl A:S93-S100).


In addition, the herein provided markers of Table 8B, Table 8C, Table 9B and Table 9C have utility for the study, identification and/or quantification of fetal cells or fetal DNA from amniocentesis and/or chorionic villus sampling. This is of particular utility in the field of prenatal diagnosis. Prenatal diagnosis procedures involve the study of fetal cells obtained by amniocentesis and chorionic villus biopsies.


Skin

The herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility for identifying individuals from traces of skin and/or adjacent tissues (such as hair, nail pieces, etc). This is of particular utility in forensic medicine and/or legal medicine. Skin or skin adjacent tissue is usually used as study material in forensic and legal medicine. The markers provided in Table 8G and 9F have a particular utility because of the following reason. Keratinocytes constitute the external layer of the skin and therefore are the first cell type to be de-attached and a high number of these cells is expected in skin traces. Variations of one marker alone or in combination with other markers herein provided or not enable the accurate assessment of identity.


In addition, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility for characterizing the skin, hair, nail, or adjacent tissue of an individual.


In addition, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility for determining the composition of the skin, hair, nail, or adjacent tissue of an individual. Said composition being dependent from the content of at least one of the three major constituting cell types of the skin (fibroblasts, keratinocytes and melanocytes).


In addition, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility in the field of drugs. They have particular utility for the development of drugs as well as for the treatment with drugs. The skin, hair, nail or adjacent tissue of an individual can be characterized by means of the provided markers of Tables 8D, G, I and Tables 9D, F, H. This information can then be used to develop new drugs or to access already existing drugs with regard to skin, hair, nail etc. of an individual or to subgroups of individuals. These subgroups are for example but not limited to be characterized by a disease and/or a defined type of skin or hair, etc. The efficiency of said drugs i.e. the presence or absence of the desired effect is also characterized or monitored by means of the provided markers of Tables 8D, G, I and Tables 9D, F, H.


In addition, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H have utility as prognostic and/or diagnostic markers for wound healing, in particular in the field of surgery procedures wherein the skin is affected.


Liver

The herein provided markers of Tables 8H and Tables 9G have utility for deducing the presence of absence of an event which affects the liver. For example but not limited to it, said event is at least one select from the group comprising liver cirrhosis; liver cancer; hepatitis A; hepatitis B; hepatitis C; healthy condition, recently or longer chemical, physical or biological exposure; recently or longer exposure to a drug, or alcohol; or treatment procedures. In the case the event is adverse, said event affecting the liver leads to the death of liver cells. In the case the event is benign, said event leads to a reduction of liver cell death. The genomic DNA of dead liver cells can then be found in the body fluids in particular in the blood of a affected individual.


In addition, the herein provided markers of Tables 8H and Tables 9G have utility for deducing the sensitivity of an individual to alcohol. Alcohol consumption may change the DNA methylation status as reviewed by Poschl et al, 2004 (Poschl G, Stickel F, Wang X D, Seitz H K. Alcohol and cancer: genetic and nutritional aspects. Proc Nutr Soc. 2004 February; 63(1):65-71.).


Heart Muscle

The herein provided markers of Tables 8E, Table 8F and Tables 9E have utility for deducing the presence of absence of an event or condition affecting the heart. For example but not limited to it, said event or condition is at least one select from the group comprising heart failure; heart attack; athletic capacity; healthy condition; recently or longer chemical, physical or biological exposure; recently or longer exposure to a drug; or treatment procedure. In the case the event is adverse, said event or condition affecting the heart leads to death of heart cells. In the case the event is benign, said event leads to a reduction of heart cell death. The genomic DNA of dead heart cells can then be found in the body fluids in particular in the blood of an affected individual.


Placenta

The herein provided markers of Table 8J and Table 9I have utility for the study, monitoring, identification and/or quantification of placental cells or placental DNA circulating in maternal blood and/or amniotic fluid. In this respect, the said markers have also utility for the isolation or purification of placental cell or placental genomic DNA. Placenta constitute an extra-embryonic fetal tissue and as such, it shares many genetic characteristics with the fetal tissue. Therefore, cells from the placenta as well as DNA from placental cells can surrogate fetal cells and fetal DNA for diagnostic means. Fetal cells and fetal DNA have a diagnostic potential in monitoring the health status of the fetus as reviewed by Bianchi D, 2004 (Bianchi D W. Circulating fetal DNA: its origin and diagnostic potential-a review. Placenta. 2004 April; 25 Suppl A:S93-S100). During pregnancy placenta cells are de-attached and brought to the maternal blood stream as well as the amniotic fluid.


In addition, the herein provided markers of Table 8J and Table 9I have utility for the monitoring of embryonic development or the monitoring of placental development, in particular of extra-embryonic tissue or of interaction of extra-embryonic tissue with maternal placental tissue.


In addition, the herein provided markers of Table 8J and Table 9I have utility for the study, monitoring, identification and/or quantification of placental cells in regenerative medicine, in particular in the field of tissue engineering. Corresponding methods for the study, monitoring, identification and/or quantification of placental cells are applied in particular before and after storage, before and after cell differentiation, before and after cell proliferation, before and after cell culture expansion, and before and after tissue expansion as well as before and after transplantation.


Sperm

The herein provided markers of Table 8L have utility for diagnosing a male infertility related disease. A major cause of male infertility is either a low amount of sperm cells (spermatozoa) in the ejaculate (oligospermia) or a complete lack of sperm cells (spermatozoa) in the ejaculate (azoospermia). Thus, methods for the quantification of sperm cells are widely useable in diagnosing male infertility.


In addition, the herein provided markers of Table 8L have utility as a tool to access the viability of the sperm cells.


In addition, the herein provided markers of Table 8L have utility for increasing the fertility of a male individual. As said above male fertility is often limited by the amount of sperm cells in the ejaculate. Thus, male fertility can be enhanced by enriching, isolating or purifying sperm cells.


In addition, the herein provided markers of Table 8L have utility for assisted fertilization procedures. Assisted fertilization procedures are for example but not limited to intracytoplasmic sperm injection (ICSI) or in vitro fertilization (IVF). All assisted fertilization procedures require the management of sperm cells prior to the procedure. Such management comprises at least the characterization, identification, quantification, enrichment, isolation, purification of sperm cells or combinations thereof.


In addition, the herein provided markers of Table 8L have utility in the fields of forensic and/or legal medicine. By use of the said markers it is possible to determine the presence or absence of sperm in a sample. Furthermore, it is possible to identify an individual by use of said markers.


Skeletal Muscle

The herein provided markers of Table 8F, 8K and Table 9J have utility for characterizing the efficiency of skeletal muscle cells. This utility is of particular value in the field of sports medicine.


In addition, the herein provided markers of Table 8F, 8K and Table 9J have utility for identifying fully differentiated muscle cells in cell culture. This is of particular utility in the field of tissue engineering. Muscle cells are generate in cell culture by cultivation and differentiation of muscle cell progenitor cells. Fully differentiated skeletal muscle cells can be identified by means of the provided markers of Table 8F, 8K and Table 9J.


In addition, the herein provided markers of Table 8F, 8K and Table 9J have utility for diagnosing muscle cell associated diseases, in particular disease which are characterized by a death of muscle cells like muscular distrophy. The DNA of dead muscle cells is found in body fluids such as blood or urine. This DNA can be identified by means of the herein provided markers of Table 8F, 8K and Table 9J.


CD8 T-Lymphocytes

The herein provided markers of Table 8A specific only for CD8 T-lymphocytes have utility for quantifying CD8 T-lymphocytes, in particular for monitoring the immune system of individuals infected with HIV. The periodically determining of the number of CD8 T-lymphocytes for patients infected with HIV is a standard procedure in the art. It is necessary to decide whether and when a drug or treatment is necessary, whether a drug or treatment is still effective, and which drug or treatment can be selected. The said is necessary with respect to the HIV infection itself but also with respect to secondary infection.


CD4+ Lymphocytes

The herein provided markers of Table 8A specific only for CD4 T-lymphocytes have utility for quantifying CD4 T-lymphocytes, in particular for monitoring the immune system of individuals infected with HIV. The periodically determining of the number of CD4 T-lymphocytes for patients infected with HIV is a standard procedure in the art. It is necessary to decide whether and when a drug or treatment is necessary, whether a drug or treatment is still effective, and which drug or treatment can be selected. The said is necessary with respect to the HIV infection itself but also with respect to secondary infection.


According to a preferred embodiment of the invention, the provided markers, in particular the genes, genomic sequences, regulatory regions, and corresponding mRNAs, cDNAs, proteins or peptides are used in the following aspects. Thereby a single marker is used either alone or in combination with other marker or markers herein provided or not.


In a preferred embodiment, at least one of the provided markers is used (i) for the characterization of the marker corresponding tissue or cell, (ii) for the identification of marker corresponding tissue or cell, (iii) for the isolation of marker corresponding tissue or cell, (iv) for the purification of the corresponding tissue or cell, or (v) combinations thereof. Therefore known methods, so far unreported methods, or combinations thereof are useable. Said application is useful in the field of research, diagnostics as well as therapeutics. As an example, but not limited to it, this application is illustrated in more detail for the marker PDGFB SEQ ID NO: 508. All the herein provided markers can be applied and used in the same way like PDGFB SEQ ID NO: 508 correspondingly to their assignment to respective tissues, organs or cells according to Tables 8 A-L or Tables 9 A-J. Of course, corresponding markers can be alternatively used. Thereby a corresponding marker of PDGFB SEQ ID NO: 508 is, for example but not limited to, genomic DNA derived from or associated with PDGFB SEQ ID NO: 508; methylation specifically converted DNA derived from PDGFB SEQ ID NO: 508; mRNA, cDNA, protein, or peptide each of which derived at least in parts from PDGFB SEQ ID NO: 508. If the case may be, a person skilled in the art knows how to adjust the presented procedure. As shown in Table 6, PDGFB SEQ ID NO: 508 is a marker for adult liver because the CpG dinucleotides of PDGFB SEQ ID NO: 508 are methylated within the range of 75-100% in liver and only within the range of 0-25% in T-lymphocytes (CD4, CD8), embryonic tissue (embryonic liver, embryonic skeletal muscle), skin (melanocytes, keratinocytes, fibroblasts), heart muscle, placenta, sperm, or skeletal muscle.


Correspondingly, for example but not limited to it, a method for characterization and/or identification of a cell or tissue type of a sample comprises the following steps:


1. Providing of a sample, the sample being derived from an individual and comprising genomic DNA. Preferably, the genomic DNA is purified by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing a cell or tissue type by determining the methylation state or the methylation level of at least one CpG position within the sequence of PDGFB SEQ ID NO: 508 of the provided sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Optional, deducing the presence or absence of a cell type or tissue type within the provided sample from the presence or absence of a methylation state or methylation level for said at least one CpG.


For example but not limited to, a method for isolation and/or purification of a cell or group of cells comprises the following steps:


1. Providing of a sample, the sample being derived from an individual and comprising one or more cells.


2. Binding of at least one probe to one or more CpG positions within the sequence of PDGFB SEQ ID NO: 508 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells.


3. Isolating and/or purifying a cell or group of cells from the provided sample by means of the attached probes and their corresponding tags, respectively. A person skilled in the art is aware of suitable methods. Said methods are based on chemical, physical or biological properties of the attached probes or corresponding tags. For example but not limited to, the isolation is performed (i) by means of affinity cromatography, wherein the probe is attached to a tag that is recognized by an antibody immobilized on a column; (ii) by means of magnetic beads, wherein a magnetic bead is directly or indirectly bound to an attached probe and wherein a magnetic field is applied; or (iii) by means of fluorescent activated cell sorting, wherein the used tag is a fluorescent dye.


In a preferred embodiment, at least one of the provided markers is used for the prospective profiling, retrospective profiling, or both of donors and/or recipients in organ transplantation procedures. The correct characterization, identification, or both of the donor and/or the recipient is mandatory during organ transplantation procedures to assure the success of the intervention. The use of the markers of the invention enables the profiling of both, donor and recipient, form which prospective or retrospective observations or conclusions about the feasibility of the procedure are drawn. As an example, but not limited to it, this application is illustrated in more detail for the marker FOXC1 SEQ ID NO: 579. All other markers of Table 8E, Table 8F and Table 9F are applied and used like FOXC1 SEQ ID NO: 579 for heart muscle. The other herein provided markers are applied and used according their assignment to the Tables 8 A-L or Tables 9 A-J. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of FOXC1 SEQ ID NO: 579 are, for example but not limited to, genomic DNA derived from or associated with FOXC1 SEQ ID NO: 579; methylation specifically converted DNA derived from FOXC1 SEQ ID NO: 579; mRNA, cDNA, protein, or peptide each of which derived at least in parts from FOXC1 SEQ ID NO: 579. If the case may be, a person skilled in the art knows how to adjust the presented procedures. As shown in Table 6, FOXC1 SEQ ID NO: 579 is a marker for heart muscle because the CpG dinucleotides of FOXC1 SEQ ID NO: 579 are methylated within the range of 25-75% in heart muscle and only within the range of 0-25% in T-lymphocytes (CD4, CD8), embryonic tissue (embryonic liver, embryonic skeletal muscle), skin (melanocytes, keratinocytes, fibroblasts), liver, placenta, sperm, or skeletal muscle.


Correspondingly, for example but not limited to it, a method for the prospective profiling, retrospective profiling, or both of a donor and/or recipient in organ or tissue transplantation procedures comprises the following steps:


1. Providing of at least one sample, at least one sample being derived from a donor individual and/or at least one sample being derived from a recipient individual. Genomic DNA is purified from said samples, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing each of the at least one sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of FOXC1 SEQ ID NO: 579 of the provided samples. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of FOXC1 SEQ ID NO: 579. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine anti-bodies.


3. Comparing the determined profiles, in particular the methylation state or methylation level of the analyzed at least one CpG position of the donor sample(s) and/or the recipient sample(s). Thereby the profiling is prospective or retrospective depending from the point in time the samples were collected i.e. before or after the transplantation procedure. A person skilled in the art knows then how to interpret the profiling to give an estimate on the probability of success of the intervention.


In a preferred embodiment, at least one of the provided markers is detected in studies by histological, chemical and/or immunohistochemical means. Said embodiment is useful in the fields of research as well as diagnostics, in particular for histological or pathological analysis. According to the said embodiment, the detection occurs by one or more probes that specifically bind to an epitop, peptide, protein, cDNA, mRNA, and/or at least one methylation state or level of at least one of the provided markers according to Tables 8 A-L or Table 9 A-J. Thereby a probe is selected from the group comprising antibody; 5-methylcytosine specific antibody (e.g. AbCam Cat. No. ab1884); affinity binding protein; protein binding specifically methylated or unmethylated DNA like MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; nucleic acid; DNA, RNA, PNA or nucleic acid derivative. In addition, the probe is labeled directly or indirectly with a dye, protein, enzyme, metal, bead or chemical compound suitable for detection. As an example, but not limited to it, this embodiment is illustrated in more detail for the marker CMAH SEQ ID NO: 570. All other markers of Table 8G and Table 9F are applied and used like CMAH SEQ ID NO: 570 for keratinocytes. The other herein provided markers are applied and used according their assignment to the Tables 8 A-L or Tables 9 A-J. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of CMAH SEQ ID NO: 570 are, for example but not limited to, genomic DNA derived from or associated with CMAH SEQ ID NO: 570; methylation specifically converted DNA derived from CMAH SEQ ID NO: 570; mRNA, cDNA, protein, or peptide each of which derived at least in parts from CMAH SEQ ID NO: 570. If the case may be, a person skilled in the art knows how to adjust the presented procedure. As shown in Table 6, CMAH SEQ ID NO: 570 is a marker for keratinocytes and sperm because the CpG dinucleotides of CMAH SEQ ID NO: 570 are methylated only within the range of 0-25% in keratinocytes and sperm and within the range of 75-100% in T-lymphocytes (CD4, CD8), embryonic tissue (embryonic liver, embryonic skeletal muscle), melanocytes, fibroblasts, heart muscle, liver, or skeletal muscle. Keratinocytes and sperm can easily be distinguished by their different morphological appearance and physiological occurrence.


Correspondingly, for example but not limited to it, a method for histological or pathological analysis, comprises


1. Providing of a sample, comprising genomic DNA;


2. Contacting the genomic DNA or a derivative of it with at least one probe which is specific for at least one differentially methylated CpG position of the marker CMAH SEQ ID NO: 570. Said probe is selected from the group comprising antibody; 5-methylcytosine specific antibody (e.g. AbCam Cat. No. ab1884); affinity binding protein; protein binding specifically methylated or unmethylated DNA like MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; nucleic acid; DNA, RNA, PNA or nucleic acid derivative specific for the methylated sequence. In addition, the probe is labeled directly or indirectly with a dye, protein, enzyme, metal, bead or chemical compound suitable for detection.


3. Performing a detection reaction by means of the probe and/or the label. A person skilled in the art is aware of suitable detection reactions. For example, but not limited to, the detection reaction comprises Rabbit Peroxidase Anti-Peroxidase (PAP) Soluble Complex (Rockland Catalog#: P300-002); radioactive labeled probes; or probes fluorescently labeled like DNA probes coupled with Cy5 (Invitrogen).


In a preferred embodiment, at least one of the provided markers is applied to phylogenetic profiling of species or tissues. The ontogenetic origin or the developmental lineage is then determined by comparison of the determined profiles. As an example, but not limited to it, this application is illustrated in more detail for the marker AIM1 SEQ ID NO: 538. All other markers of Table 8J and Table 9I are applied and used like AIM1 SEQ ID NO: 538 for placenta. The other herein provided markers are applied and used according their assignment to the Tables 8 A-L or Tables 9 A-J. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of AIM1 SEQ ID NO: 538 are, for example but not limited to, genomic DNA derived from or associated with AIM1 SEQ ID NO: 538; methylation specifically converted DNA derived from AIM1 SEQ ID NO: 538; mRNA, cDNA, protein, or peptide each of which derived at least in parts AIM1 SEQ ID NO: 538. If the case may be, a person skilled in the art knows how to adjust the presented procedures. As shown in Table 6, AIM1 SEQ ID NO: 538 is a marker for placenta because the CpG dinucleotides of AIM1 SEQ ID NO: 538 are methylated within the range of 25-75% in placenta and only within the range of 0-25% in T-lymphocytes (CD4, CD8), embryonic tissue (embryonic liver, embryonic skeletal muscle), skin (melanocytes, keratinocytes, fibroblasts), liver, heart muscle, sperm, or skeletal muscle.


Correspondingly, for example but not limited to it, a method for phylogenetic profiling of species or tissues, comprises


1. Providing of at least one sample, each sample comprising genomic DNA. Genomic DNA is purified from said samples, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing each of the at least one sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of AIM1 SEQ ID NO: 538 of the provided samples. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of AIM1 SEQ ID NO: 538. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine anti-bodies.


The profiles of the one or more respective samples are then compared with each other or with at least one reference profile. According to methods or algorithms known to those skilled in the art, the ontogenetic origin of a cell, group of cells, tissue or organ or the developmental lineage of a cell, group of cells, tissue or organ is determined.


In a preferred embodiment, at least one of the provided markers is applied for quality control of a genetically modified organism, tissue, group of cells or cell. As an example, but not limited to it, this embodiment is illustrated in more detail for the marker TBC1D10A SEQ ID NO: 700. All other markers of Table 8E, 8F and Table 9E are applied and used like TBC1D10A SEQ ID NO: 700 for heart muscle. The other herein provided markers are applied and used according their assignment to the Tables 8 A-L or Tables 9 A-J. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of TBC1D10A SEQ ID NO: 700 are, for example but not limited to, genomic DNA derived from or associated with TBC1D10A SEQ ID NO: 700; methylation specifically converted DNA derived from TBC1D10A SEQ ID NO: 700; mRNA, cDNA, protein, or peptide each of which derived at least in parts from TBC1D10A SEQ ID NO: 700. If the case may be, a person skilled in the art knows how to adjust the presented procedures. TBC1D10A SEQ ID NO: 700 is a marker for heart muscle because a) the CpG dinucleotides of TBC1D10A SEQ ID NO: 700 are methylated to a significantly higher extend in the heart muscle and sperm than in other tissues (see Table 6); and b) heart muscle and sperm can be easily distinguished morphologically or by means of other markers. According to Table 6 75-100% of the CpG dinucleotides of the marker TBC1D10A SEQ ID NO: 700 is methylated in heart muscle and sperm; 0-25% is methylated in T-lymphocytes (CD4, CD8), embryonic tissue (embryonic liver, embryonic skeletal muscle), skin (melanocytes, keratinocytes, fibroblasts), and placenta; and 25-75% is methylated in liver and skeletal muscle.


Correspondingly, for example but not limited to it, a method for quality control of a genetically modified organism, tissue, group of cells or cell, comprises


1. Providing of at least one sample of or derived from the genetically modified organism, tissue, group of cells or cell, each sample comprising genomic DNA. The genomic DNA is purified from said samples, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing each of the at least one sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of TBC1D10A SEQ ID NO: 700 of the provided samples. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of TBC1D10A SEQ ID NO: 700. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine anti-bodies.


3. Comparing the said profiles with each other or with reference profiles. A person skilled in the art knows to interpret correspondingly said comparison and to deduce therefrom the quality of the genetically modified organism, tissue, group of cells or cell. Thereby a person skilled in the art is enabled to draw prospectively or retrospectively conclusions on the presence or absence of side effects if said genetically modified organism, tissue, group of cells or cell is brought into contact with other organisms, tissues, groups of cells or cells.


For example but not limited to, a method for quality control of an genetically modified organism, tissue, group of cells or cell, comprises


1. Providing of at least one first sample of or derived from an organism, tissue, group of cells or cell being not genetically modified and at least one second sample of or derived from a correspondent organism, tissue, group of cells or cell being genetically modified. Thereby each sample comprises genomic DNA. The genomic DNA is purified from said first and second samples, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing each of the at least one first and second samples by determining the methylation state or the methylation level of at least one CpG position within the sequence of TBC1D10A SEQ ID NO: 700 of the provided samples. Thereby a first and second profile is generated comprising the methylation information of all characterized CpG positions of TBC1D10A SEQ ID NO: 700 of the respective samples. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine anti-bodies.


3. Comparing the said first and second profile with each other or with reference profiles.


4. Deducing the presence or absence of side effects, wherein said side effects are characterized in that changes are introduced into regions of the genome which were not target of the genetic modification. This application is of particular interest to exclude unwanted physiological alterations of the cell, group of cells, tissue or organism. The reason for this is unwanted physiological alterations are mainly caused by genetic modification of non-target genomic DNA regions.


According to a particular preferred embodiment, the at least one first sample is derived from an organism, tissue, group of cells or cell before a genetic modification and the at least one second sample is derived thereof after said genetic modification.


In a preferred embodiment, at least one of the provided markers is applied for controlling side effects in in vivo gene therapy procedures wherein genetically modified organism, tissue, group of cells or cell is used. As an example, but not limited to it, this embodiment is illustrated in more detail for the marker GPX5 SEQ ID NO: 574. All other markers of Table 8A and Table 9A are applied and used correspondingly as GPX5 SEQ ID NO: 574 for T-lymphocytes. The other herein provided markers are applied and used according their assignment to the Tables 8 A-L or Tables 9 A-J. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of GPX5 SEQ ID NO: 574 are, for example but not limited to, genomic DNA derived from or associated with GPX5 SEQ ID NO: 574; methylation specifically converted DNA derived from GPX5 SEQ ID NO: 574; mRNA, cDNA, protein, or peptide each of which derived at least in parts from GPX5 SEQ ID NO: 574. If the case may be, a person skilled in the art knows how to adjust the presented procedures. As shown in Table 6, GPX5 SEQ ID NO: 574 is a marker for T-lymphocytes because the CpG dinucleotides of GPX5 SEQ ID NO: 574 are methylated within the range of 0-25% in CD4 T-lymphocytes as well as in CD 8 T-lymphocytes and within the range of 75-100% in embryonic tissue (embryonic liver, embryonic skeletal muscle), skin (melanocytes, keratinocytes, fibroblasts), placenta, liver, heart muscle, sperm, or skeletal muscle.


For example but not limited to, a method for controlling side effects in in vivo gene therapy procedures, comprises


1. Providing of at least one untreated sample derived from an individual and at least one treated sample of said individual. Thereby the samples are derived from respective body regions and each of the samples comprises genomic DNA. The genomic DNA is purified from said first and second samples, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing each of the at least one first and second samples by determining the methylation state or the methylation level of at least one CpG position within the sequence of GPX5 SEQ ID NO: 574 of the provided samples. Thereby a first and second profile is generated comprising the methylation information of all characterized CpG positions of GPX5 SEQ ID NO: 574 of the respective samples. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine anti-bodies.


3. Comparing the said first and second profile with each other or with reference profiles.


4. Deducing the presence or absence of side effects, wherein said side effects are characterized in that changes are introduced into regions of the genome which were not target of the genetic modification. This application is of particular interest to exclude unwanted physiological alterations for the individual. The reason for this unwanted physiological alterations are mainly caused by genetic modification of non-target genomic DNA regions.


According to a particular preferred embodiment, the at least one untreated sample is derived before the gene therapy and the at least one treated sample is derived after said gene therapy.


In a preferred embodiment, at least one of the provided markers is applied for the characterization, identification, or labelling of corresponding tissue or combinations thereof. Said embodiment is of particular use in the field of tissue bank storage and proliferation. Furthermore it can be used in a prospective as well as in a retrospective manner. The provided markers allow the individualization of samples by a precise molecular method. This is mandatory in storing biological material from patients or healthy individuals. In addition, this also advantageous for isolation or purification of tissues cells. As an example, but not limited to it, this application is illustrated in more detail for the marker TCN2 SEQ ID NO: 470. All other markers of Table 8L are applied and used like TCN2 SEQ ID NO: 470 for sperm. The other herein provided markers are applied and used according their assignment to the Tables 8 A-L or Tables 9 A-J. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of TCN2 SEQ ID NO: 470 are, for example but not limited to, genomic DNA derived from or associated with TCN2 SEQ ID NO: 470; methylation specifically converted DNA derived from TCN2 SEQ ID NO: 470; mRNA, cDNA, protein, or peptide each of which derived at least in parts from TCN2 SEQ ID NO: 470. If the case may be, a person skilled in the art knows how to adjust the presented procedures. As shown in Table 6, TCN2 SEQ ID NO: 470 is a marker for sperm because the CpG dinucleotides of TCN2 SEQ ID NO: 470 are methylated within the range of 75-100% in sperm and to only an extend of 0-25% in T-lymphocytes (CD4, CD8), embryonic tissue (embryonic liver, embryonic skeletal muscle), skin (melanocytes, keratinocytes, fibroblasts), placenta, liver, heart muscle, or skeletal muscle.


Correspondingly, for example but not limited to it, a method for characterizing a tissue or cell, comprises


1. Providing of at least one sample comprising genomic DNA. The genomic DNA is purified from said sample(s), preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing each sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of TCN2 SEQ ID NO: 470 of the provided sample(s). Thereby a profile is generated comprising the methylation information of all characterized CpG positions of TCN2 SEQ ID NO: 470 of the respective sample(s). A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


For example but not limited to it, a method for labelling a tissue or cell, comprises in addition to the said method for characterizing a tissue or cell:


3. Labelling said tissue or cell by means of the methylation status or level of one or more of the analyzed CpG positions of TCN2 SEQ ID NO: 470. According to a preferred embodiment the labelling is achieved by assigning at least one distinct methylation status or level of at least one analyzed CpG dinucleotide of TCN2 SEQ ID NO: 470 to said tissue or cell. Thereby the assigned methylation status or level(s) are specific for said tissue or cell.


For example but not limited to it, a method for identifying a tissue or cell, comprises in addition to the said method for characterizing a tissue or cell:


3. Identifying a tissue or cell or an individual from whom the sample is derived from by comparison of the determined TCN2 profile of said samples with a reference TCN2 profile.


For example but not limited to it, a method for profiling a tissue type or cell type, comprises in addition to the said method for characterizing a tissue or cell:


3. Comparing the determined TCN2 profiles of said samples with each other and/or with at least one reference TCN2 profile and considering the group of same methylation status or levels of correspondent CpG dinucleotides of different samples as a tissue type or cell type profile.


A person skilled in the art knows to combine the said methods for characterizing a tissue or cell; for labelling a tissue or cell; for identifying a tissue or cell; and for profiling a tissue type or cell type with state of the art methods for tissue or cell isolation or purification.


In a preferred embodiment, at least one of the provided markers is applied for controlling cell differentiation in stem-cell research and/or therapeutics. Cells undergo many genetic and/or epigenetic changes throughout differentiation. These changes influence the physiology of the cell and their control is mandatory in any procedure involving stem-cell in research and/or therapeutics. The provided markers enable to control this changes by giving a reference of the adult (completely differentiated) and embryonic (partially differentiated) status of the cells. As an example, but not limited to it, this application is illustrated in more detail for the marker RPL3 SEQ ID NO: 529. All other markers of Table 8H and Table 9G are applied and used like RPL3 SEQ ID NO: 529 for adult liver. The other herein provided markers are applied and used according their assignment to the Tables 8 A-L or Tables 9 A-J. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of RPL3 SEQ ID NO: 529 are, for example but not limited to, genomic DNA derived from or associated with RPL3 SEQ ID NO: 529; methylation specifically converted DNA derived from RPL3 SEQ ID NO: 529; mRNA, cDNA, protein, or peptide each of which derived at least in parts from RPL3 SEQ ID NO: 529. If the case may be, a person skilled in the art knows how to adjust the presented procedures. As shown in Table 6, RPL3 SEQ ID NO: 529 is a marker for sperm because the CpG dinucleotides of RPL3 SEQ ID NO: 529 are methylated within the range of 25-75% in adult liver and within the range of 75-100% in T-lymphocytes (CD4, CD8), embryonic tissue (embryonic liver, embryonic skeletal muscle), skin (melanocytes, keratinocytes, fibroblasts), placenta, sperm, heart muscle, or skeletal muscle.


Correspondingly, for example but not limited to it, a method for controlling cell differentiation in stem-cell research and/or therapeutics, comprises


1. Providing of a sample comprising genomic DNA. The genomic DNA is purified from said sample(s), preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing said sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of RPL3 SEQ ID NO: 529 of the provided sample. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of RPL3 SEQ ID NO: 529 of the respective sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Comparing said RPL3 profile of said sample with at least one reference RPL3 profile and deducing therefrom if a desired differentiation status of one or more cells of the sample is met or not. Through this a controlling of cell differentiation is achieved.


According to Table 6, the herein provided markers are assigned to different tissues (see Tables 8 A-L or Tables 9 A-J). In the following, applications are described which are only applicable for the named tissue(s).


CD4+ and CD8+ Lymphocytes:

In a preferred embodiment, the herein provided markers of Table 8A and Table 9A are used for the quantification of lymphocytes, in particular in peripheral blood.


Low number of leucocytes in blood (leucopenia) may indicate:

    • bone marrow failure (for example, due to infection, tumor, fibrosis),
    • presence of cytotoxic substance,
    • collagen-vascular diseases (such as lupus erythematosus),
    • disease of the liver or spleen,
    • exposure to radiation,


High number of leucocytes in blood (leucocytosis) may indicate:

    • infectious diseases,
    • inflammatory disease (such as rheumatoid arthritis or allergy),
    • leukemia,
    • severe emotional or physical stress,
    • tissue damage (for example, burns),
    • anemia.


Said markers enable the identification of CD4+ and CD8+ lymphocytes among other cells in blood samples. For example but not limited to it, the differential methylation of FBLN1 SEQ ID NO: 426 is used. According to Table 6, the differential methylation of FBLN1 SEQ ID NO: 426 is marker for CD4 T-lymphocytes as well as for CD8 T-lymphocytes because the CpG dinucleotides of FBLN1 SEQ ID NO: 426 are methylated within the range of 25-75% in CD4 and CD8 T-lymphocytes while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of FBLN1 SEQ ID NO: 426 are, for example but not limited to, genomic DNA derived from or associated with FBLN1 SEQ ID NO: 426; methylation specifically converted DNA derived from FBLN1 SEQ ID NO: 426; mRNA, cDNA, protein, or peptide each of which derived at least in parts from FBLN1 SEQ ID NO: 426. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to it, a method for the quantification of CD4 and/or CD8 T-lymphocytes, comprises:


1. Providing of a sample, comprising genomic DNA;


2. Contacting the genomic DNA or a derivative of it with at least one probe which is specific for at least one differentially methylated CpG position of the marker FBLN1 SEQ ID NO: 426. Said probe is selected from the group comprising antibody; 5-methylcytosine specific antibody (e.g. AbCam Cat. No. ab1884); affinity binding protein; protein binding specifically methylated or unmethylated DNA like MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; nucleic acid; DNA, RNA, PNA or nucleic acid derivative specific for the methylated sequence. In addition, the probe is labeled directly or indirectly with a dye, protein, enzyme, metal, bead or chemical compound suitable for detection.


3. Performing a detection reaction by means of the probe and/or the label. A person skilled in the art is aware of suitable detection reactions. For example, but not limited to, the detection reaction comprises Rabbit Peroxidase Anti-Peroxidase (PAP) Soluble Complex (Rockland Catalog#: P300-002); radioactive labeled probes; or probes fluorescently labeled like DNA probes coupled with Cy5 (Invitrogen). A person skilled in the art knows further suitable methods for detection.


4. Quantifying the detection reaction in a manner so that the detected signal is indicative for the amount of probe or label therewith also for the amount of CD4 and/or CD8 T-lymphocytes. A person knows suitable methods for quantification.


In a particular preferred embodiment, the said method is also a method for isolation of CD4 and CD8 T-lymphocytes. Said method additional comprises the separation of CD4 and CD8 T-lymphocytes from other cells, tissue, or molecules of a sample by means of the said probes and/or labels attached to one or more differentially methylated CpG position of FBLN1 SEQ ID NO: 426 in CD4 and CD8 T-lymphocytes. A person skilled in the art knows suitable methods for separation of labeled cells form unlabeled cells, tissue, or molecules.


Alternatively, for example but not limited to, a method for quantifying the number of CD4 and/or CD8 T-lymphocytes comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of FBLN1 SEQ ID NO: 426 of the provided sample. A person skilled in the art knows how to determine the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of CD4 and/or CD8 T-lymphocytes by comparing the determined one or more methylation levels of the sample with the respective herein provided one or more methylation levels of CD4 and/or CD8 T-lymphocytes and with at least one methylation level representing the correspondent tissue, group of cells, or cell of a healthy individual.


In a preferred embodiment, the markers of Table 8A and Table 9A are used to study the CD4 and/or CD8 T-lymphocyte infiltration in other tissues healthy or diseased. Infiltration of lymphocytes in healthy or diseased tissues is an indication of several diseases such immunological malignances or even in tumor development. The said markers represent a target for the development of molecular probes that coupled to any detection method (e.g. Fluorescent dye) allow the identification of these cells in histological preparations. For example but not limited to it, the differential methylation of HLA-DPB SEQ ID NO: 416 is used. According to Table 6, the differential methylation of HLA-DPB SEQ ID NO: 416 is marker for CD4 T-lymphocytes as well as for CD8 T-lymphocytes because the CpG dinucleotides of HLA-DPB SEQ ID NO: 416 are methylated within the range of 75-100% in CD4 and CD8 T-lymphocytes while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of HLA-DPB SEQ ID NO: 416 are, for example but not limited to, genomic DNA derived from or associated with HLA-DPB SEQ ID NO: 416; methylation specifically converted DNA derived from HLA-DPB SEQ ID NO: 416; mRNA, cDNA, protein, or peptide each of which derived at least in parts from HLA-DPB SEQ ID NO: 416. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to it, a method for detection of infiltrated CD4 and/or CD8 T-lymphocytes comprises:

    • 1. Providing of a sample, comprising genomic DNA;
    • 2. Contacting the genomic DNA or a derivative of it with at least one probe which is specific for at least one differentially methylated CpG position of the marker HLA-DPB SEQ ID NO: 416. Said probe is selected from the group comprising antibody; 5-methylcytosine specific antibody (e.g. AbCam Cat. No. ab1884); affinity binding protein; protein binding specifically methylated or unmethylated DNA like MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; nucleic acid; DNA, RNA, PNA or nucleic acid derivative specific for the methylated sequence. In addition, the probe is labeled directly or indirectly with a dye, protein, enzyme, metal, bead or chemical compound suitable for detection.
    • 3. Performing a detection reaction by means of the probe and/or the label. A person skilled in the art knows of suitable detection reactions. For example, but not limited to, the detection reaction comprises Rabbit Peroxidase Anti-Peroxidase (PAP) Soluble Complex (Rockland Catalog#: P300-002); radioactive labeled probes; or probes fluorescently labeled like DNA probes coupled with Cy5 (Invitrogen).


According to a preferred embodiment, said method is performed in a histological manner. A person skilled in the art knows how to carried such a method. For example, but not limited to, the providing of the sample comprises the making of sample sections suitable for histological analysis.


In a preferred embodiment, the markers of Table 8A and Table 9A are used to identify, isolate and/or purify CD4 T-lymphocytes and/or CD8 T-lymphocytes, in particular from surrounding tissue infiltrated by the T-lymphocytes; from blood; and/or from other body fluids. For example but not limited to it, the differential methylation of APOBEC3B SEQ ID NO: 474 is used. According to Table 6, the differential methylation of APOBEC3B SEQ ID NO: 474 is a marker for CD4 T-lymphocytes as well as for CD8 T-lymphocytes because the CpG dinucleotides of APOBEC3B SEQ ID NO: 474 are methylated within the range of 0-25% in CD4 and CD8 T-lymphocytes while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of APOBEC3B SEQ ID NO: 474 are, for example but not limited to, genomic DNA derived from or associated with APOBEC3B SEQ ID NO: 474; methylation specifically converted DNA derived from APOBEC3B SEQ ID NO: 474; mRNA, cDNA, protein, or peptide each of which derived at least in parts from APOBEC3B SEQ ID NO: 474. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for identifying CD4 T-lymphocytes and/or CD8 T-lymphocytes comprises:


1. Providing of a sample, the sample comprising genomic DNA.


2. Binding of at least one probe to one or more CpG positions within the sequence of APOBEC3B SEQ ID NO: 474 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells. A person skilled in the art knows suitable methods to carry out this step.


3. Identifying CD4 and/or CD8 T-lymphocytes by detecting the bound probes and/or their respective label(s). A person skilled in the art knows suitable methods for detection of said probes or labels.


For example but not limited to, a method for isolating and/or purifying CD4 and/or CD8 T-lymphocytes comprises in addition to the steps of the said method for identifying CD4 T-lymphocytes and/or CD8 T-lymphocytes:


3. Isolating and/or purifying of the identified CD4 and/or CD8 T-lymphocytes from the provided sample by means of the attached probes and their corresponding tags, respectively. A person skilled in the art knows suitable methods. Said methods are based on chemical, physical or biological properties of the attached probes or corresponding tags. For example but not limited to, the isolation is performed (i) by means of affinity cromatography, wherein the probe is attached to a tag that is recognized by an antibody immobilized on a column; (ii) by means of magnetic beads, wherein a magnetic bead is directly or indirectly bound to an attached probe and wherein a magnetic field is applied; or (iii) by means of fluorescent activated cell sorting, wherein the used tag is a fluorescent dye.


According to a preferred embodiment, the isolated or purified CD4 and/or CD8 T-lymphocytes are quantified by means of the attached probes and/or their corresponding tags. A person skilled in the art knows suitable methods. For example, but not limited to by cell counting manually or by automatic means.


In a preferred embodiment, the markers of Table 8A and Table 9A are used for the identification of an individual. Thereby at least two samples are used. One samples is obtained from an individual and another sample is a forensic sample, in particular traces of body cells, tissues or fluids for example but not limited to traces of blood and/or body fluids. This embodiment is of particular use in the field of forensic medicine or of legal medicine. As constituent of blood or body fluids, CD4 T-lymphocytes and CD8 T-lymphocytes are part of the mentioned traces. The said markers have the advantage of being stable over time because they are DNA based. In addition said markers have the advantage that they enable a highly detailed and accurate characterization of samples. Through this an unambiguous identification of an individual is enabled. For example but not limited to it, the differential methylation of GPX5 SEQ ID NO: 574 is used. According to Table 6, the differential methylation of GPX5 SEQ ID NO: 574 is a marker for CD4 T-lymphocytes as well as for CD8 T-lymphocytes because the CpG dinucleotides of GPX5 SEQ ID NO: 574 are methylated within the range of 0-25% in CD4 and CD8 T-lymphocytes while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of GPX5 SEQ ID NO: 574 are, for example but not limited to, genomic DNA derived from or associated with GPX5 SEQ ID NO: 574; methylation specifically converted DNA derived from GPX5 SEQ ID NO: 574; mRNA, cDNA, protein, or peptide each of which derived at least in parts from GPX5 SEQ ID NO: 574. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to it, a method for the identification of an individual, comprises


1. Providing at least two samples. One sample is collected from an individual. Another sample is a forensic sample. Each of the provided samples comprises genomic DNA. The genomic DNA is purified from said samples, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing each sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of GPX5 SEQ ID NO: 574 of the provided samples. Thereby a profile is generated comprising the methylation information of all characterized CpG positions GPX5 SEQ ID NO: 574 of the respective samples. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3.Comparing the GPX5 profile of the forensic sample with one or more profiles of the samples collected from individual. An individual is identified wherein the GXP5 profile matches the profile of the sample of said individual. In alternative preferred embodiments, the forensic sample and the samples collected from individuals are collected or processed not at the same time.


In a preferred embodiment, the markers of Table 8A and Table 9A are used to diagnose the presence or absence of a disease. Thereby the number of CD4 T-lymphocytes, CD8 T-lymphocytes or both is quantified in normalized samples of healthy individuals. The determined number of CD4 T-lymphocytes, CD8 T-lymphocytes or both are then considered as indicative for healthy condition or a diseased condition with respect to an individual. Preferably, large amount of normalized samples are considered to generate reference values of CD4 T-lymphocytes, CD8 T-lymphocytes or both for a healthy condition and/or for one or more diseased conditions. The diseased condition can be any kind of diseased condition. Preferably, the diseased condition is a disease which causes a immune reaction. For example but not limited to the diseased condition is a cancer disease, a cell proliferation disease, or HIV. Preferably the total number of cells present in a sample is determined. The number of CD4 T-lymphocytes, CD8 T-lymphocytes or both are then normalized to the total number of cells. For example but not limited to it, the differential methylation of SYNE1 SEQ ID NO: 558 is used. According to Table 6, the differential methylation of SYNE1 SEQ ID NO: 558 is a marker for CD4 T-lymphocytes, for CD8 T-lymphocytes as well as sperm because the CpG dinucleotides of SYNE1 SEQ ID NO: 558 are methylated within the range of 75-100% in CD4 T-lymphocytes, CD8 T-lymphocytes and sperm while other tissues show a different extend of methylation. Because sperm can be easily morphologically distinguished from CD4 or CD8 T-lymphocytes, this marker can be used in the following described application. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of SYNE1 SEQ ID NO: 558 are, for example but not limited to, genomic DNA derived from or associated with SYNE1 SEQ ID NO: 558; methylation specifically converted DNA derived from SYNE1 SEQ ID NO: 558; mRNA, cDNA, protein, or peptide each of which derived at least in parts from SYNE1 SEQ ID NO: 558. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to it, a method for diagnosing the absence or presence of a diseased or healthy condition for an individual, comprises

    • 1. Providing a sample, comprising genomic DNA.
    • 2. Determining the number of CD4 T-lymphocytes, CD8 T-lymphocytes or both by detecting the methylation status or level of at least one CpG position within the sequence of SYNE1 SEQ ID NO: 558 of the provided samples. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.
    • 3. Normalizing the determined number of CD4 T-lymphocytes, CD8 T-lymphocytes or CD4 T-lymphocytes and CD8 T-lymphocytes. A person skilled in the art knows suitable methods for normalization. For example but not limited to the determined number of CD4 T-lymphocytes, CD8 T-lymphocytes or CD4 T-lymphocyte and CD8 T-lymphocytes is normalized to the number of total cell contained the provided sample.


      1. In a preferred embodiment, step 2 is performed in a histological, immunohistological and/or immunocytological manners. A person skilled in the art knows suitable methods for carrying out step 2 in histological, immunohistological and/or immunocytological manners for example but not limited to in situ hybridization, antibody stainings, or FACS. In another preferred embodiment, step 2 is performed by means of molecular biological methods. A person skilled in the art knows also suitable methods therefor for example but not limited to real time PCR based methods or hybridization based methods (Microarrays).


Embryonic

In a preferred embodiment, the herein provided markers of Table 8B, Table 8C, Table 9B and Table 9C are used for the study, identification and/or quantification of fetal cells or fetal DNA circulating in maternal blood and/or amniotic fluid. During pregnancy cells and DNA from the fetus are continuously brought to the maternal blood stream as well as the amniotic fluid. Fetal cells and fetal DNA have a diagnostic potential in monitoring the health status of the fetus as reviewed by Bianchi D, 2004 (Bianchi D W. Circulating fetal DNA: its origin and diagnostic potential-a review. Placenta. 2004 April; 25 Suppl A:S93-S100). For example but not limited to it, the differential methylation of CTA-384D8.15 SEQ ID NO: 509 is used. According to Table 6, the differential methylation of CTA-384D8.15 SEQ ID NO: 509 is a marker for embryonic liver because the CpG dinucleotides of CTA-384D8.15 SEQ ID NO: 509 are methylated within the range of 25-75% in embryonic liver while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of CTA-384D8.15 SEQ ID NO: 509 are, for example but not limited to, genomic DNA derived from or associated with CTA-384D8.15 SEQ ID NO: 509; methylation specifically converted DNA derived from CTA-384D8.15 SEQ ID NO: 509; mRNA, cDNA, protein, or peptide each of which derived at least in parts from CTA-384D8.15 SEQ ID NO: 509. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for identifying fetal cells or fetal DNA comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA.


2. Binding of at least one probe to one or more CpG positions within the sequence of CTA-384D8.15 SEQ ID NO: 509 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells. A person skilled in the art knows suitable methods to carry out this step.


3. Identifying fetal cells or fetal genomic DNA by detecting the bound probes and/or their respective label(s). A person skilled in the art knows suitable methods for detection of said probes or labels.


In preferred embodiments, a probe of step 2 binds specifically with respect to the methylation status of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 22, 25, or 30 CpG dinucleotides.


For example but not limited to, a method for isolating and/or purifying fetal cells or fetal genomic DNA comprises in addition to the steps of the said method for identifying fetal cells or fetal genomic DNA:


3. Isolating and/or purifying of the identified fetal cells or fetal genomic DNA from the provided sample by means of the attached probes and their corresponding tags, respectively. A person skilled in the art knows suitable methods. Said methods are based on chemical, physical or biological properties of the attached probes or corresponding tags. For example but not limited to, the isolation is performed (i) by means of affinity cromatography, wherein the probe (e.g. a nucleic acid) is directly or indirectly bound to a solid surface; (ii) by means of affinity cromatography, wherein the probe is attached to a tag that is recognized by an antibody immobilized on a column; (iii) by means of magnetic beads, wherein a magnetic bead is directly or indirectly bound to an attached probe and wherein a magnetic field is applied; or (iv) by means of fluorescent activated cell sorting, wherein the used tag is a fluorescent dye.


According to a preferred embodiment, the isolated or purified fetal cells or fetal genomic DNA are quantified by means of the attached probes and/or their corresponding tags. A person skilled in the art knows suitable methods. For example, but not limited to by cell counting manually or by automatic means. According to a preferred embodiment, the isolated or purified fetal genomic DNA is quantified by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method.


Alternatively, for example but not limited to, a method for quantifying the number of fetal cells or the amount of fetal genomic DNA comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of CTA-384D8.15 SEQ ID NO: 509 of the provided sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of fetal cells or the amount of fetal genomic DNA by comparing the determined one or more methylation levels of the sample with the respective herein provided one or more methylation levels specific for embryonic liver and/or embryonic skeletal muscle and with at least one methylation level representing the correspondent tissue, group of cells, or cell comprising no placental DNA.


For example but not limited to it, a method for characterizing one or more fetal cells or fetal genomic DNA comprises


1. Providing of a sample comprising one or more fetal cells or fetal genomic DNA. The fetal cell(s) or the fetal genomic DNA are isolated for example but not limited to the methods described herein.


2. Characterizing said sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of CTA-384D8.15 SEQ ID NO: 509 of the provided sample. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of CTA-384D8.15 SEQ ID NO: 509 of the respective sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


In a preferred embodiment of step 1, the genomic DNA comprising fetal genomic DNA is isolated/purified from the provided sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega). In a preferred embodiment the determined CTA-384D8.15 profile is compared with one or more CTA-384D8.15 profiles of obtained from other samples and/or with one or more CTA-384D8.15 reference profiles.


In a preferred embodiment, the herein provided markers of Table 8B, Table 8C, Table 9B and Table 9C are used for the study, identification and/or quantification of fetal cells or fetal DNA from amniocentesis and/or chorionic villus sampling. Said embodiment is of particular use in the field of prenatal diagnosis. Prenatal diagnosis procedures involve the study of fetal cells obtained by amniocentesis and chorionic villus biopsies. For example but not limited to it, the differential methylation of CRYBA4 SEQ ID NO: 476 is used. According to Table 6, the differential methylation of CRYBA4 SEQ ID NO: 476 is a marker for embryonic liver and embryonic skeletal muscle because the CpG dinucleotides of CRYBA4 SEQ ID NO: 476 are methylated within the range of 25-75% in embryonic liver or embryonic skeletal muscle while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of CRYBA4 SEQ ID NO: 476 are, for example but not limited to, genomic DNA derived from or associated with CRYBA4 SEQ ID NO: 476; methylation specifically converted DNA derived from CRYBA4 SEQ ID NO: 476; mRNA, cDNA, protein, or peptide each of which derived at least in parts from CRYBA4 SEQ ID NO: 476. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for identifying fetal cells or fetal DNA comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA.


2. Binding of at least one probe to one or more CpG positions within the sequence of CRYBA4 SEQ ID NO: 476 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells. A person skilled in the art knows suitable methods to carry out this step.


3. Identifying fetal cells or fetal genomic DNA by detecting the bound probes and/or their respective label(s). A person skilled in the art knows suitable methods for detection of said probes or labels.


In preferred embodiments, a probe of step 2 binds specifically with respect to the methylation status of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 22, 25, or 30 CpG dinucleotides.


For example but not limited to, a method for isolating and/or purifying fetal cells or fetal genomic DNA comprises in addition to the steps of the said method for identifying fetal cells or fetal genomic DNA:


3. Isolating and/or purifying of the identified fetal cells or fetal genomic DNA from the provided sample by means of the attached probes and their corresponding tags, respectively. A person skilled in the art knows suitable methods. Said methods are based on chemical, physical or biological properties of the attached probes or corresponding tags. For example but not limited to, the isolation is performed (i) by means of affinity cromatography, wherein the probe (e.g. a nucleic acid) is directly or indirectly bound to a solid surface; (ii) by means of affinity cromatography, wherein the probe is attached to a tag that is recognized by an antibody immobilized on a column; (iii) by means of magnetic beads, wherein a magnetic bead is directly or indirectly bound to an attached probe and wherein a magnetic field is applied; or (iv) by means of fluorescent activated cell sorting, wherein the used tag is a fluorescent dye.


According to a preferred embodiment, the isolated or purified fetal cells or fetal genomic DNA are quantified by means of the attached probes and/or their corresponding tags. A person skilled in the art knows suitable methods. For example, but not limited to by cell counting manually or by automatic means. According to a preferred embodiment, the isolated or purified fetal genomic DNA is quantified by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method.


Alternatively, for example but not limited to, a method for quantifying the number of fetal cells or the amount of fetal genomic DNA comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of CRYBA4 SEQ ID NO: 476 of the provided sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of fetal cells or the amount of fetal genomic DNA by comparing the determined methylation levels of the sample with the respective herein provided methylation levels of embryonic liver and/or embryonic skeletal muscle.


For example but not limited to it, a method for characterizing one or more fetal cells or fetal genomic DNA comprises


1. Providing of a sample comprising one or more fetal cells or fetal genomic DNA. The fetal cell(s) or the fetal genomic DNA are isolated for example but not limited to the methods described herein.


2. Characterizing said sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of CRYBA4 SEQ ID NO: 476 of the provided sample. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of CRYBA4 SEQ ID NO: 476 of the respective sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


In a preferred embodiment of step 1, the genomic DNA comprising fetal genomic DNA is isolated/purified from the provided sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


In a preferred embodiment the determined CRYBA4 profile is compared with one or more CRYBA4 profiles of obtained from other samples and/or with one or more CRYBA4 reference profiles.


Skin:

In a preferred embodiment, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H are used for identifying individuals from traces of skin and/or adjacent tissues (such as hair, nail pieces, etc). This embodiment is of particular use in forensic medicine and/or legal medicine. Skin or skin adjacent tissue is usually used as study material in forensic and legal medicine. Preferably the markers provided in Table 8G and 9F are used because of the following reason. Keratinocytes constitute the external layer of the skin and therefore are the first cell type to be de-attached and a high number of these cells is expected in skin traces. Variations of one marker alone or in combination with other markers herein provided or not enable the accurate assessment of identity. For example but not limited to it, the differential methylation of NP612444.1 SEQ ID NO: 689 is used. According to Table 6, the differential methylation of NP612444.1 SEQ ID NO: 689 is a marker for keratinocytes as well as for sperm because the CpG dinucleotides of NP612444.1 SEQ ID NO: 689 are methylated within the range of 0-25% in keratinocytes and sperm while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of NP612444.1 SEQ ID NO: 689 are, for example but not limited to, genomic DNA derived from or associated with NP612444.1 SEQ ID NO: 689; methylation specifically converted DNA derived from NP612444.1 SEQ ID NO: 689; mRNA, cDNA, protein, or peptide each of which derived at least in parts from NP612444.1 SEQ ID NO: 689. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to it, a method for the identification of an individual, comprises


1. Providing a sample, comprising skin, hair, nail pieces and/or adjacent tissue. Genomic DNA is purified from said samples, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing the sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of NP612444.1 SEQ ID NO: 689 of the provided sample. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of NP612444.1 SEQ ID NO: 689 of the sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3.Comparing the determined NP612444.1 profile of the sample with one or more NP612444.1 profiles of individuals, the profiles obtained correspondingly or in a different manner. An individual is identified wherein the NP612444.1 profile matches the profile of an individual. In preferred embodiments, the forensic sample and the sample collected from an individual are collected and/or processed simultaneously or not.


In a preferred embodiment, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H are used to characterize the skin, hair, nail, or adjacent tissue of an individual. For example but not limited to it, the differential methylation of SEQ ID NO: 640 (no gene associated) is used. According to Table 6, the differential methylation of SEQ ID NO: 640 is a marker for skin, hair, nail, or adjacent tissue because the CpG dinucleotides of SEQ ID NO: 640 are methylated within the range of 25-75% in keratinocytes, melanocytes and fibroblasts while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of SEQ ID NO: 640 are, for example but not limited to, genomic DNA derived from or associated with SEQ ID NO: 640; methylation specifically converted DNA derived from SEQ ID NO: 640; mRNA, cDNA, protein, or peptide each of which derived at least in parts from SEQ ID NO: 640. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for characterizing skin, hair, nail and/or adjacent tissue comprises the characterization of at least keratinocytes, melanocytes, fibroblasts or combinations thereof, in addition comprising:


1. Providing a sample, comprising skin, hair, nail and/or adjacent tissue. Genomic DNA is purified from said samples, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing the sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of SEQ ID NO: 640 of the provided sample. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of SEQ ID NO: 640 of the sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


In a preferred embodiment, the herein provided markers of Tables 8D, G, 1 and Tables 9D, F, H are used to determine the composition of the skin, hair, nail, or adjacent tissue of an individual. Said composition being dependent from the content of at least one of the three major constituting cell types of the skin (fibroblasts, keratinocytes and melanocytes). For example but not limited to it, the differential methylation of SEQ ID NO: 644 (no gene associated), SEQ ID NO: 648 (no gene associated), PTPNS1 SEQ ID NO: 649, or combinations thereof are used. According to Table 6, the differential methylation of SEQ ID NO: 644 is a marker for melanocytes because the CpG dinucleotides of SEQ ID NO: 644 are methylated within the range of 0-25% in melanocytes while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of SEQ ID NO: 644 are, for example but not limited to, genomic DNA derived from or associated with SEQ ID NO: 644; methylation specifically converted DNA derived from SEQ ID NO: 644; mRNA, cDNA, protein, or peptide each of which derived at least in parts from SEQ ID NO: 644. According to Table 6, the differential methylation of SEQ ID NO: 648 is a marker for fibroblasts because the CpG dinucleotides of SEQ ID NO: 648 are methylated within the range of 0-25% in fibroblast while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of SEQ ID NO: 648 are, for example but not limited to, genomic DNA derived from or associated with SEQ ID NO: 648; methylation specifically converted DNA derived from SEQ ID NO: 648; mRNA, cDNA, protein, or peptide each of which derived at least in parts from SEQ ID NO: 648. According to Table 6, the differential methylation of PTPNS1 SEQ ID NO: 649 is a marker for keratinocytes because the CpG dinucleotides of PTPNS1 SEQ ID NO: 649 are methylated within the range of 0-25% in keratinocytes while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of PTPNS1 SEQ ID NO: 649 are, for example but not limited to, genomic DNA derived from or associated with PTPNS1 SEQ ID NO: 649; methylation specifically converted DNA derived from PTPNS1 SEQ ID NO: 649; mRNA, cDNA, protein, or peptide each of which derived at least in parts from PTPNS1 SEQ ID NO: 649. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for quantifying the number of keratinocytes, fibroblast, melanocytes or combinations thereof comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA.


2. Binding of at least one probe to one or more CpG positions within the sequence of SEQ ID NO: 644, SEQ ID NO: 648, PTPNS1 SEQ ID NO: 649 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells. A person skilled in the art knows suitable methods to carry out this step.


3. Identifying keratinocytes, fibroblasts, melanocytes or combinations thereof by detecting the bound probes and/or their respective label(s). A person skilled in the art knows suitable methods for detection of said probes or labels.


4. Quantifying the isolated or purified cells by means of the attached probes and/or their corresponding tags. A person skilled in the art knows suitable methods. For example, but not limited to by cell counting manually or by automatic means. According to a preferred embodiment, the isolated or purified fetal genomic DNA is quantified by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method.


Alternatively, for example but not limited to, a method for quantifying the number of keratinocytes, fibroblast, melanocytes or combinations thereof comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of SEQ ID NO: 644, SEQ ID NO: 648, PTPNS1 SEQ ID NO: 649 or combinations thereof of the provided sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of keratinocytes, fibroblasts, melanocytes, or combinations thereof by comparing the determined methylation levels of the sample with the respective herein provided methylation levels of keratinocytes, fibroblasts or melanocytes.


In a preferred embodiment, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H are used in the field of drugs. Said embodiment is of particular use for the development of drugs as well as for the treatment with drugs. The skin, hair, nail or adjacent tissue of an individual can be characterized by means of the provided markers of Tables 8D, 6, I and Tables 9D, F, H. This information can then be used to develop new drugs or to access already existing drugs with regard to skin, hair, nail etc. of an individual or to subgroups of individuals. These subgroups are for example but not limited to be characterized by a disease and/or a defined type of skin or hair, etc. The efficiency of said drugs i.e. the presence or absence of the desired effect is also characterized or monitored by means of the provided markers of Tables 8D, G, I and Tables 9D, F, H. For example but not limited to it, the differential methylation of SEQ ID NO: 773 (no gene associated) is used. According to Table 6, the differential methylation of SEQ ID NO: 773 is a marker for skin, hair, nail, or adjacent tissue because the CpG dinucleotides of SEQ ID NO: 773 are methylated within the range of 0-25% in keratinocytes, melanocytes and fibroblasts while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of SEQ ID NO: 773 are, for example but not limited to, genomic DNA derived from or associated with SEQ ID NO: 773; methylation specifically converted DNA derived from SEQ ID NO: 773; mRNA, cDNA, protein, or peptide each of which derived at least in parts from SEQ ID NO: 773. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for developing a drug and/or for treating an individual with a drug comprises:


1. Providing a sample obtained from an individual comprising genomic DNA of keratinocytes, melanocytes, fibroblasts or combinations thereof. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing said sample by determining the methylation status or level of at least one CpG position within the sequence of SEQ ID NO: 773 of the provided sample. Thereby a SEQ ID NO: 773 profile specific for said sample is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Treating said individual with a drug.


4. Providing a sample of said individual after drug treatment.


5. Characterizing said sample after drug treatment by determining the methylation status or level of at least one CpG position within the sequence of SEQ ID NO: 773 of the provided sample. Thereby a SEQ ID NO: 773 profile specific for said sample after drug treatment is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


6. Comparing the determined SEQ ID NO: 773 profiles with drug treatment and without drug treatment with each other.


According to a preferred embodiment, guidelines are drawn from the comparison for the further drug development. According to another preferred embodiment, guidelines are drawn from the comparison for the treatment of an individual with said drug.


In a preferred embodiment, the herein provided markers of Tables 8D, G, I and Tables 9D, F, H are used as prognostic and/or diagnostic markers for wound healing, in particular in the field of surgery procedures wherein the skin is affected. For example but not limited to it, the differential methylation of SLC35E4 SEQ ID NO: 751 is used. According to Table 6, the differential methylation of SLC35E4 SEQ ID NO: 751 is a marker for wound healing because the CpG dinucleotides of SLC35E4 SEQ ID NO: 751 are methylated within the range of 0-25% in keratinocytes, melanocytes and fibroblasts while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of SLC35E4 SEQ ID NO: 751 are, for example but not limited to, genomic DNA derived from or associated with SLC35E4 SEQ ID NO: 751; methylation specifically converted DNA derived from SLC35E4 SEQ ID NO: 751; mRNA, cDNA, protein, or peptide each of which derived at least in parts from SLC35E4 SEQ ID NO: 751. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for prognosing or diagnosing would healing comprises:


1. Providing a sample obtained from an individual comprising genomic DNA of keratinocytes, melanocytes, fibroblasts or combinations thereof. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing said sample by determining the methylation status or level of at least one CpG position within the sequence of SLC35E4 SEQ ID NO: 751 of the provided sample. Thereby a SLC35E4 profile specific for said sample is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Comparing the determined SLC35E4 profile with at least one reference profile obtained from the same individual and/or from one or more other individuals. Preferably, said reference profiles are obtained according to steps 1 and 2. Preferably, said reference profiles comprises pairs of profiles each pair being obtained from an individual. Thereby said pair consists of a first profile specific for the condition of healthy skin (no lesion of skin) and of a second profile specific for a healing state of the wound or affected skin area after lesion. A person skilled in the art knows to deduce the possible grade of scare building and or the time required for healing therefrom.


In a preferred embodiment, said method is a method for diagnosing wound healing wherein the determined SLC35E4 profile matches a reference profile. In another preferred embodiment, said method is a method for prognosing wound healing wherein the determined SLC35E4 profile matches a first reference profile and wherein a second reference profile exists which was obtained from the same individual as the first reference profile. Thereby the second reference profile was obtained after the first reference profile. Accordingly, the prognosis for the individual for whom the SLC35E4 profile was determined is the condition of wound healing characterized by the second reference profile. In preferred embodiments, at least 1, 2, 4, 6, 10, 15, 25 or 50 reference profiles are considered. The more reference profiles are considered the merrier is the diagnosis or prognosis.


Liver

In a preferred embodiment, the herein provided markers of Tables 8H and Tables 9G are used for deducing the presence of absence of an event which affects the liver. For example but not limited to it, said event is at least one select from the group comprising liver cirrhosis; liver cancer; hepatitis A; hepatitis B; hepatitis C; healthy condition, recently or longer chemical, physical or biological exposure; recently or longer exposure to a drug, or alcohol; or treatment procedures. In the case the event is adverse, said event affecting the liver leads to the death of liver cells. In the case the event is benign, said event leads to a reduction of liver cell death. The genomic DNA of dead liver cells can then be found in the body fluids in particular in the blood of a affected individual. As an example but not limited to, the differential methylation of VARS SEQ ID NO: 415 is used. According to Table 6, the differential methylation of VARS SEQ ID NO: 415 is a marker for diseases affecting the liver because the CpG dinucleotides of VARS SEQ ID NO: 415 are methylated within the range of 25-75% in liver while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of VARS SEQ ID NO: 415 are, for example but not limited to, genomic DNA derived from or associated with VARS SEQ ID NO: 415; methylation specifically converted DNA derived from VARS SEQ ID NO: 415; mRNA, cDNA, protein, or peptide each of which derived at least in parts from VARS SEQ ID NO: 415. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for detecting a liver affecting event comprises:


1. Providing a sample comprising genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega). Preferably said sample is a blood sample, plasma sample, or urine sample.


2. Determining the methylation level of at least one CpG position within the sequence of VARS SEQ ID NO: 415 of the provided sample. Thereby a VARS profile specific of said sample is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Deducing the presence or absence of a liver affecting event from the comparison of the determined VARS profile with one or more VARS reference profiles. Said reference profiles are specific for a healthy condition or a condition specific for an event. In the case the determined VARS profile matches or is similar to a reference profile specific for an event, said liver affecting event is present. In case the determined VARS profiles is not similar to a reference profile specific for an event, said liver affecting event is absent.


In addition, for example but not limited to it, a method for detecting a liver affecting event comprises the quantification of the amount of free floating genomic DNA of liver cells. Said method comprises:

    • 1. Providing of a sample, comprising genomic DNA;
    • 2. Contacting the genomic DNA or a derivative of it with at least one probe which is specific for at least one differentially methylated CpG position of the marker VARS SEQ ID NO: 415. Said probe is selected from the group comprising antibody; 5-methylcytosine specific antibody (e.g. AbCam Cat. No. ab1884); affinity binding protein; protein binding specifically methylated or unmethylated DNA like MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; nucleic acid; DNA, RNA, PNA or nucleic acid derivative specific for the methylated sequence. In addition, the probe is labeled directly or indirectly with a dye, protein, enzyme, metal, bead or chemical compound suitable for detection.
    • 3. Performing a detection reaction by means of the probe and/or the label. A person skilled in the art is aware of suitable detection reactions. For example, but not limited to, the detection reaction comprises Rabbit Peroxidase Anti-Peroxidase (PAP) Soluble Complex (Rockland Catalog#: P300-002); radioactive labeled probes; or probes fluorescently labeled like DNA probes coupled with Cy5 (Invitrogen). A person skilled in the art knows further suitable methods for detection.
    • 4. Quantifying the detection reaction in a manner so that the detected signal is indicative for the amount of probe or label and therewith for the number of dead liver cell. A person knows suitable methods for quantification.
    • 5. Deducing the presence or absence of a liver affecting event from the comparison of the determined number of dead liver cells with reference numbers of dead liver cells that are specific for an event. In the case the determined number matches or is similar to a reference number specific for a healthy condition, a liver affecting event is absent. In the case the determined number matches or is similar to a reference number specific for a condition specific for an event, a liver affecting event is present.


Alternatively, for example but not limited to, a method for detecting a liver affecting event comprises the quantification of the amount of free floating genomic DNA of liver cells. Said method comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of VARS SEQ ID NO: 415 of the provided sample. A person skilled in the art knows how to determine the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of dead liver cells by comparing the determined one or more methylation levels of the sample with the respective herein provided one or more methylation levels of liver cells and with at least one methylation level representing the correspondent tissue, group of cells, or cell of a healthy individual.


In a preferred embodiment, the herein provided markers of Tables 8H and Tables 9G are used for deducing the sensitivity of an individual to alcohol. Alcohol consumption changes the DNA methylation status as reviewed by Poschl et al, 2004 (Poschl G, Stickel F, Wang X D, Seitz H K. Alcohol and cancer: genetic and nutritional aspects. Proc Nutr Soc. 2004 February; 63(1):65-71.). As an example but not limited to, the differential methylation of BMP7 SEQ ID NO: 684 is used. According to Table 6, the differential methylation of BMP7 SEQ ID NO: 684 is a marker for diseases affecting the liver because the CpG dinucleotides of BMP7 SEQ ID NO: 684 are methylated within the range of 25-75% in liver while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of BMP7 SEQ ID NO: 684 are, for example but not limited to, genomic DNA derived from or associated with BMP7 SEQ ID NO: 684; methylation specifically converted DNA derived from BMP7 SEQ ID NO: 684; mRNA, cDNA, protein, or peptide each of which derived at least in parts from BMP7 SEQ ID NO: 684. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for deducing the sensitivity of an individual to alcohol comprises:


1. Providing a sample derived from said individual comprising genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega). Preferably said sample is a blood sample, plasma sample, or urine sample.


2. Determining the methylation level of at least one CpG position within the sequence of BMP7 SEQ ID NO: 684 of the provided sample. Thereby a BMP7 profile specific of said sample is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Deducing the extend of a sensitivity of said individual to alcohol from the comparison of the determined BMP7 profile with one or more BMP7 reference profiles. The reference profiles are specific for different conditions of sensitivity. In the case the determined BMP7 profile matches or is similar to a reference profile, the sensitivity of said individual is identical or similar to the sensitivity for which the reference profile is specific for.


In a preferred embodiment, the sensitivity to alcohol is determined for a cell, group of cell, or tissue according to the above described procedure.


Heart Muscle:

In a preferred embodiment, the herein provided markers of Tables 8E, Table 8F and Tables 9E are used for deducing the presence of absence of an event or condition affecting the heart. For example but not limited to it, said event or condition is at least one select from the group comprising heart failure; heart attack; athletic capacity; healthy condition; recently or longer chemical, physical or biological exposure; recently or longer exposure to a drug; or treatment procedure. In the case the event is adverse, said event or condition affecting the heart leads to death of heart cells. In the case the event is benign, said event leads to a reduction of heart cell death. The genomic DNA of dead heart cells can then be found in the body fluids in particular in the blood of an affected individual. As an example but not limited to, the differential methylation of TBC1D10A SEQ ID NO: 700 is used. According to Table 6, the differential methylation of TBC1D10A SEQ ID NO: 700 is a marker for diseases affecting the heart because the CpG dinucleotides of TBC1D10A SEQ ID NO: 700 are methylated within the range of 75-100% in heart while other tissues show a different extend of methylation. Of course, corresponding markers can also be used alternatively. Thereby corresponding markers of TBC1D10A SEQ ID NO: 700 are, for example but not limited to, genomic DNA derived from or associated with TBC1D10A SEQ ID NO: 700; methylation specifically converted DNA derived from TBC1D10A SEQ ID NO: 700; mRNA, cDNA, protein, or peptide each of which derived at least in parts from TBC1D10A SEQ ID NO: 700. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for detecting a heart affecting event or condition comprises:


1. Providing a sample comprising genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega). Preferably said sample is a blood sample, plasma sample, or urine sample.


2. Determining the methylation level of at least one CpG position within the sequence of TBC1D10A SEQ ID NO: 700 of the provided sample. Thereby a TBC1D10A profile specific of said sample is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Deducing the presence or absence of a heart affecting event or condition from the comparison of the determined TBC1D10A profile with one or more TBC1D10A reference profiles. Said reference profiles are specific for a healthy condition or a condition specific for an event. In case the determined TBC1D10A profile matches or is similar to a reference profile specific for an event or condition, the said heart affecting event or condition is present. In case the determined TBC1D10A profile is not similar to a reference profile specific for an event or condition, the said heart affecting event or condition is absent.


In addition, for example but not limited to it, a method for detecting a heart affecting event comprises the quantification of the amount of free floating genomic DNA of heart cells. Said method comprises:

    • 1. Providing of a sample, comprising genomic DNA;
    • 2. Contacting the genomic DNA or a derivative of it with at least one probe which is specific for at least one differentially methylated CpG position of the marker TBC1D10A SEQ ID NO: 700. Said probe is selected from the group comprising antibody; 5-methylcytosine specific antibody (e.g. AbCam Cat. No. ab1884); affinity binding protein; protein binding specifically methylated or unmethylated DNA like MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; nucleic acid; DNA, RNA, PNA or nucleic acid derivative specific for the methylated sequence. In addition, the probe is labeled directly or indirectly with a dye, protein, enzyme, metal, bead or chemical compound suitable for detection.
    • 3. Performing a detection reaction by means of the probe and/or the label. A person skilled in the art is aware of suitable detection reactions. For example, but not limited to, the detection reaction comprises Rabbit Peroxidase Anti-Peroxidase (PAP) Soluble Complex (Rockland Catalog#: P300-002); radioactive labeled probes; or probes fluorescently labeled like DNA probes coupled with Cy5 (Invitrogen). A person skilled in the art knows further suitable methods for detection.
    • 4. Quantifying the detection reaction in a manner so that the detected signal is indicative for the amount of probe or label and therewith for the number of dead liver cell. A person knows suitable methods for quantification.
    • 5. Deducing the presence or absence of a heart affecting event from the comparison of the determined number of dead heart cells with reference numbers of dead heart cells that are specific for an event. In the case the determined number matches or is similar to a reference number specific for an event or condition, said heart affecting event is present. In the case the determined number is not similar to a reference number specific for an event or condition, said heart affecting event is absent.


Alternatively, for example but not limited to, a method for detecting a heart affecting event comprises the quantification of the amount of free floating genomic DNA of heart cells. Said method comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of TBC1D10A SEQ ID NO: 700 of the provided sample. A person skilled in the art knows how to determine the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of dead heart cells by comparing the determined one or more methylation levels of the sample with the respective herein provided one or more methylation levels of heart cells and with at least one methylation level representing the correspondent tissue, group of cells, or cell of a healthy individual.


Placenta:

In a preferred embodiment, the herein provided markers of Table 8J and Table 9I are used for the study, monitoring, identification and/or quantification of placental cells or placental DNA circulating in maternal blood and/or amniotic fluid. Placenta constitute an extra-embryonic fetal tissue and as such, it shares many genetic characteristics with the fetal tissue. Therefore, cells from the placenta as well as DNA from placental cells can surrogate fetal cells and fetal DNA for diagnostic means. Fetal cells and fetal DNA have a diagnostic potential in monitoring the health status of the fetus as reviewed by Bianchi D, 2004 (Bianchi D W. Circulating fetal DNA: its origin and diagnostic potential-a review. Placenta. 2004 April; 25 Suppl A:S93-S100). During pregnancy placenta cells are de-attached and brought to the maternal blood stream as well as the amniotic fluid. For example but not limited to it, the differential methylation of PRAME SEQ ID NO: 419 is used. According to Table 6, the differential methylation of PRAME SEQ ID NO: 419 is a marker for placenta because the CpG dinucleotides of PRAME SEQ ID NO: 419 are methylated within the range of 0-25% in placenta while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of PRAME SEQ ID NO: 419 are, for example but not limited to, genomic DNA derived from or associated with PRAME SEQ ID NO: 419; methylation specifically converted DNA derived from PRAME SEQ ID NO: 419; mRNA, cDNA, protein, or peptide each of which derived at least in parts from PRAME SEQ ID NO: 419. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for identifying one or more placental cells or placental DNA comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA.


2. Binding of at least one probe to one or more CpG positions within the sequence of PRAME SEQ ID NO: 419 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells. A person skilled in the art knows suitable methods to carry out this step.


3. Identifying placental cells or placental genomic DNA by detecting the bound probes and/or their respective label(s). A person skilled in the art knows suitable methods for detection of said probes or labels.


In preferred embodiments, a probe of step 2 binds specifically with respect to the methylation status of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 22, 25, or 30 CpG dinucleotides.


For particular applications it is necessary to isolate or purify placental cell or placental genomic DNA. Accordingly, as an example but not limited to, a method for isolating and/or purifying placental cells or placental genomic DNA comprises in addition to the steps of the said method for identifying placental cells or placental genomic DNA:


3. Isolating and/or purifying of the identified placental cells or placental genomic DNA from the provided sample by means of the attached probes and their corresponding tags, respectively. A person skilled in the art knows suitable methods. Said methods are based on chemical, physical or biological properties of the attached probes or corresponding tags. For example but not limited to, the isolation is performed (i) by means of affinity cromatography, wherein the probe (e.g. a nucleic acid) is directly or indirectly bound to a solid surface; (ii) by means of affinity cromatography, wherein the probe is attached to a tag that is recognized by an antibody immobilized on a column; (iii) by means of magnetic beads, wherein a magnetic bead is directly or indirectly bound to an attached probe and wherein a magnetic field is applied; or (iv) by means of fluorescent activated cell sorting, wherein the used tag is a fluorescent dye.


According to a preferred embodiment, the isolated or purified placental cells or placental genomic DNA are quantified by means of the attached probes and/or their corresponding tags. A person skilled in the art knows suitable methods. For example, but not limited to by cell counting manually or by automatic means. According to a preferred embodiment, the isolated or purified placental genomic DNA is quantified by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method.


Alternatively, for example but not limited to, a method for quantifying the number of placental cells or the amount of placental genomic DNA comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of PRAME SEQ ID NO: 419 of the provided sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of placental cells or the amount of placental genomic DNA by comparing the determined one or more methylation levels of the sample with the respective herein provided one or more methylation levels specific for placenta and with at least one methylation level representing the correspondent tissue, group of cells, or cell comprising no placental DNA.


For example but not limited to it, a method for characterizing one or more placental cells or placental genomic DNA comprises


1. Providing of a sample comprising one or more placental cells or placental genomic DNA. The placental cell(s) or the placental genomic DNA are isolated for example but not limited to the methods described herein.


2. Characterizing said sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of PRAME SEQ ID NO: 419 of the provided sample. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of PRAME SEQ ID NO: 419 of the respective sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


In a preferred embodiment of step 1, the genomic DNA comprising placental genomic DNA is isolated/purified from the provided sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


In a preferred embodiment, the determined PRAME profile is compared with one or more PRAME profiles obtained from other samples of the same individual and/or with one or more PRAME reference profiles of a normal pregnancy.


In a preferred embodiment, the herein provided markers of Table 8J and Table 9I are used for to the monitoring of embryonic development or the monitoring of placental development, in particular of extra-embryonic tissue or of interaction of extra-embryonic tissue with maternal placental tissue. Said embodiment comprises one or more of said methods for identifying one or more placental cells or placental DNA; one or more of said methods for isolating and/or purifying one or more placental cells or placental genomic DNA; one or more of said methods for quantifying the number of placental cells or the amount of placental genomic DNA; one or more of said methods for characterizing one or more placental cells or placental genomic DNA; or combinations thereof. application is for example but not limited


In a preferred embodiment, the herein provided markers of Table 8J and Table 9I are used for the study, monitoring, identification and/or quantification of placental cells in regenerative medicine, in particular in the field of tissue engineering. Therefore the above described methods (a method for identifying one or more placental cells or placental DNA; a method for isolating and/or purifying one or more placental cells or placental genomic DNA; a method for quantifying the number of placental cells or the amount of placental genomic DNA; a method for characterizing one or more placental cells or placental genomic DNA; a method for monitoring one or more placental cells) or combinations thereof are used. Thereby the said methods are applied to placental cells or to cells derived from placental cells. Furthermore the methods are applied in particular before and after storage, before and after cell differentiation, before and after cell proliferation, before and after cell culture expansion, and before and after tissue expansion as well as before and after transplantation. For example but not limited to, the differential methylation of GPR24 SEQ ID NO: 436 is used. According to Table 6, the differential methylation of GPR24 SEQ ID NO: 436 is a marker for placenta because the CpG dinucleotides of GPR24 SEQ ID NO: 436 are methylated within the range of 25-75% in placenta while other tissues show a different extend of methylation. Said methods (a method for identifying one or more placental cells or placental DNA; a method for isolating and/or purifying one or more placental cells or placental genomic DNA; a method for quantifying the number of placental cells or the amount of placental genomic DNA; a method for characterizing one or more placental cells or placental genomic DNA; a method for monitoring one or more placental cells) are carried out as described above, wherein PRAME SEQ ID NO: 419 is substituted by GPR24 SEQ ID NO: 436. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of GPR24 SEQ ID NO: 436 are, for example but not limited to, genomic DNA derived from or associated with GPR24 SEQ ID NO: 436; methylation specifically converted DNA derived from GPR24 SEQ ID NO: 436; mRNA, cDNA, protein, or peptide each of which derived at least in parts from GPR24 SEQ ID NO: 436. If the case may be, a person skilled in the art knows how to adjust the said methods.


Sperm:

The herein provided markers of Table 8L are used for diagnosing a male infertility related disease. A major cause of male infertility is either a low amount of sperm cells (spermatozoa) in the ejaculate (oligospermia) or a complete lack of sperm cells (spermatozoa) in the ejaculate (azoospermia). Thus, methods for the quantification of sperm cells are widely used in diagnosis of male infertility. In addition methylation analysis of sperm cells can be used as a tool to access the viability of the said. For example but not limited to, the differential methylation of GAL3ST1 SEQ ID NO: 437 is used. According to Table 6, the differential methylation of GAL3ST1 SEQ ID NO: 437 is a marker for sperm because the CpG dinucleotides of GAL3ST1 SEQ ID NO: 437 are methylated within the range of 0-25% in sperm while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of GAL3ST1 SEQ ID NO: 437 are, for example but not limited to, genomic DNA derived from or associated with GAL3ST1 SEQ ID NO: 437; methylation specifically converted DNA derived from GAL3ST1 SEQ ID NO: 437; mRNA, cDNA, protein, or peptide each of which derived at least in parts from GAL3ST1 SEQ ID NO: 437. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for diagnosing a male infertility related disease comprises the quantification and/or characterization of sperm cells in the ejaculate.


For example, but not limited to, a method for characterization of sperm cells in the ejaculate comprises:


1. Providing of a sample comprising ejaculate or genomic DNA derived from an ejaculate. The genomic DNA is isolated/purified from the provided sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing said sample by determining the methylation state or the methylation level of at least one CpG position within the sequence of GAL3ST1 SEQ ID NO: 437 of the provided sample. Thereby a profile is generated comprising the methylation information of all characterized CpG positions of GAL3ST1 SEQ ID NO: 437 of the respective sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


According to a preferred embodiment, the determined GAL3ST1 profile of a provided sample is compared with at least one GAL3ST1 reference profile. Said reference profile is either obtained from a different individual or from the same individual. The reference profile is characterized in that it is specific for a defined state of viability of sperm cells. In a preferred embodiment several reference profiles are used each of which is specific for a defined state of viability of sperm cells in the ejaculate and therefore for a defined value of infertility or fertility. In case a determined GAL3ST1 profile matches or is similar to a reference profile, it is deduced that the viability of sperm cells of the correspondent individual is characterized by the said defined state of viability of the reference.


Alternatively, for example, but not limited to, a method for characterizing sperm cells in an ejaculate comprises:

    • 1. Providing of a sample comprising ejaculate or genomic DNA derived from an ejaculate.
    • 2. Binding of at least one probe to one or more CpG positions within the sequence of GAL3ST1 SEQ ID NO: 437 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions creating a characteristic binding pattern. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof, or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells. A person skilled in the art knows suitable methods to carry out this step.


According to a preferred embodiment, the determined GAL3ST1 binding pattern is compared with at least one GAL3ST1 reference binding pattern. Said reference binding pattern is either obtained from a different individual or from the same individual. The reference binding pattern is characterized in that it is specific for a defined state of viability of sperm cells. In a preferred embodiment several reference binding patterns are used each of which is specific for a defined state of viability of sperm cells in the ejaculate and therefore for a defined value of infertility or fertility. In case a determined GAL3ST1 binding pattern matches or is similar to a reference binding pattern, it is deduced that the viability of sperm cells of the correspondent individual is characterized by the said defined state of viability of the reference.


For example, but not limited to, a method for quantification of sperm cells in the ejaculate comprises the identification of sperm cells. For example, but not limited to, a method for identification of sperm cells comprises:


1. Providing of a sample comprising ejaculate or genomic DNA derived from an ejaculate.


2. Binding of at least one probe to one or more CpG positions within the sequence of GAL3ST1 SEQ ID NO: 437 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions creating a characteristic binding pattern. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells. A person skilled in the art knows suitable methods to carry out this step.


3. Identifying sperm cells or sperm genomic DNA by detecting the bound probes and/or their respective label(s). A person skilled in the art knows suitable methods for detection of said probes or labels.


In preferred embodiments, a probe of step 2 binds specifically with respect to the methylation status of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 22, 25, or 30 CpG dinucleotides.


The said method for quantification of sperm cells in the ejaculate comprising the identification of sperm cells additional comprises:


Quantifying the identified sperm cells by counting or quantifying the labeled sperm genomic DNA by quantifying the bound probes and/or their respective label(s). A person skilled in the art knows suitable methods for quantification and/or counting.


Alternatively, for example but not limited to, a method for quantification of sperm cells in the ejaculate comprises:


1. Providing of a sample comprising ejaculate or genomic DNA derived from an ejaculate. The genomic DNA is isolated/purified from the provided sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of GAL3ST1 SEQ ID NO: 437 of the provided sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of sperm cells or the amount of sperm genomic DNA by comparing the determined one or more methylation levels of the sample with the respective herein provided one or more methylation levels specific for sperm and with at least one methylation level representing an ejaculate comprising no sperm cell or a defined amount of sperm cells.


For example but not limited to, a method for diagnosing fertility or infertility for a male individual comprises:

    • 1. Quantifying the amount of sperm cells in the ejaculate of said individual.
    • 2. Comparing said amount with at least one reference value. The reference value is specific for a defined fertility or infertility state. In case the quantified amount of sperm cells matches or is similar to the reference value, it is deduced that the fertility or infertility of said individual has the same fertility or infertility which is specific for the reference value.


In a preferred embodiment, the herein provided markers of Table 8L are used for increasing the fertility of a male individual. As said above male fertility is often limited by the amount of sperm cells in the ejaculate. Thus, male fertility can be enhanced by enriching, isolating or purifying sperm cells. For example but not limited to, the differential methylation of APOL4 SEQ ID NO: 486 is used. According to Table 6, the differential methylation of APOL4 SEQ ID NO: 486 is a marker for sperm because the CpG dinucleotides of APOL4 SEQ ID NO: 486 are methylated within the range of 75-100% in sperm while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of APOL4 SEQ ID NO: 486 are, for example but not limited to, genomic DNA derived from or associated with APOL4 SEQ ID NO: 486; methylation specifically converted DNA derived from APOL4 SEQ ID NO: 486; mRNA, cDNA, protein, or peptide each of which derived at least in parts from APOL4 SEQ ID NO: 486. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for increasing the fertility of a male individual comprises:


1. Identification of sperm cells in an ejaculate by means of the above described method for identification of sperm cells, wherein GAL3ST1 SEQ ID NO: 437 is substitutes by APOL4 SEQ ID NO: 486.


2. Enriching, isolating or purifying sperm cells by means of the attached probes and their corresponding tags, respectively. A person skilled in the art knows suitable methods. Said methods are based on chemical, physical or biological properties of the attached probes or corresponding tags. For example but not limited to, the isolation is performed (i) by means of affinity cromatography, wherein the probe (e.g. a nucleic acid) is directly or indirectly bound to a solid surface; (ii) by means of affinity cromatography, wherein the probe is attached to a tag that is recognized by an antibody immobilized on a column; (iii) by means of magnetic beads, wherein a magnetic bead is directly or indirectly bound to an attached probe and wherein a magnetic field is applied; or (iv) by means of fluorescent activated cell sorting, wherein the used tag is a fluorescent dye.


In a preferred embodiment, the herein provided markers of Table 8L are used for assisted fertilization procedures. Assisted fertilization procedures are for example but not limited to intracytoplasmic sperm injection (ICSI) or in vitro fertilization (IVF). All assisted fertilization procedures require the management of sperm cells prior to the procedure. Such management comprises at least the characterization, identification, quantification, enrichment, isolation, purification of sperm cells or combinations thereof. Therefore the above described methods for characterizing, identifying, quantifying, enriching, isolating and purifying sperm cells are applied.


In a preferred embodiment, the herein provided markers of Table 8L are used in the fields of forensic and/or legal medicine. By use of the said markers it is possible to determine the presence or absence of sperm in a sample. Furthermore, it is possible to identify an individual by use of said markers. For example but not limited to, the differential methylation of TCN2 SEQ ID NO: 470 is used. According to Table 6, the differential methylation of TCN2 SEQ ID NO: 470 is a marker for sperm because the CpG dinucleotides of TCN2 SEQ ID NO: 470 are methylated within the range of 75-100% in sperm while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of TCN2 SEQ ID NO: 470 are, for example but not limited to, genomic DNA derived from or associated with TCN2 SEQ ID NO: 470; methylation specifically converted DNA derived from TCN2 SEQ ID NO: 470; mRNA, cDNA, protein, or peptide each of which derived at least in parts from TCN2 SEQ ID NO: 470. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for identifying sperm cells in a sample and/or for identifying an individual comprises:

    • 1. Providing a sample comprising genomic DNA.
    • 2. Binding of at least one probe to one or more CpG positions within the sequence of TCN2 SEQ ID NO: 470 of the provided sample. Thereby a probe binds specifically with respect to the methylation status of said one or more CpG positions and/or with respect to the methylation status of said one or more CpG positions and an individual. A probe is either a protein, peptide, nucleic acid, RNA or DNA for example but not limited to, an antibody specific for 5-methylcytosine (e.g. AbCAM Cat. No. ab1884); a methyl-binding protein such as the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof, or a nucleic acid probe that is specific for the methylated sequence. According to some preferred embodiments, the said probe(s) are labeled with a tag suitable for detection of the probe, isolation of one or more cells, and/or purification of one or more cells. A person skilled in the art knows suitable methods to carry out this step.
    • 3. Identifying sperm cells or sperm genomic DNA or identifying sperm cells or sperm genomic DNA of an individual by detecting the bound probes and/or their respective label(s). A person skilled in the art knows suitable methods for detection of said probes or labels.


In addition, for example but not limited to, an alternative method for identifying an individual comprises:


1. Providing of a sample comprising genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of TCN2 SEQ ID NO: 470 of the provided sample. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Comparing the determined methylation level(s) of said sample with respective TCN2 SEQ ID NO: 470 methylation level(s) of at least one sample obtained from at least one candidate individual. Wherein the determined methylation level(s) matches or is similar to the respective methylation level(s) of an individual, it its deduced that the provided sample comprises sperm genomic DNA or sperm cells of said individual. Thereby an individual is identified.


Skeletal Muscle

In a preferred embodiment, the herein provided markers of Table 8F, 8K and Table 9J are used for characterizing the efficiency of skeletal muscle cells. This embodiment is of particular value in the field of sports medicine. For example but not limited to, the differential methylation of CARD10 SEQ ID NO: 498 is used. According to Table 6, the differential methylation of CARD10 SEQ ID NO: 498 is a marker for skeletal muscle because the CpG dinucleotides of CARD10 SEQ ID NO: 498 are methylated within the range of 0-25% in skeletal muscle while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of CARD10 SEQ ID NO: 498 are, for example but not limited to, genomic DNA derived from or associated with CARD10 SEQ ID NO: 498; methylation specifically converted DNA derived from CARD10 SEQ ID NO: 498; mRNA, cDNA, protein, or peptide each of which derived at least in parts from CARD10 SEQ ID NO: 498. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for characterizing the efficiency of a skeletal muscle comprises:


1. Providing of a sample comprising genomic DNA of a skeletal muscle. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing the provided sample by Determining the methylation level of at least one CpG position within the sequence of CARD10 SEQ ID NO: 498 of the provided sample. Thereby a CARD 10 profile is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Comparing the generated CARD10 profile with at least one CARD10 reference profile. Thereby each reference profile is characteristic for a defined efficiency. Wherein the determined CARD10 profile matches or is similar to a CARD10 reference profile, the skeletal muscle from whom the analyzed sample is provided has the same efficiency as the said reference.


In a preferred embodiment, the herein provided markers of Table 8F, 8K and Table 9J are used for identifying fully differentiated muscle cells in cell culture. This is of particular value in the field of tissue engineering. Muscle cells are generate in cell culture by cultivation and differentiation of muscle cell progenitor cells. Fully differentiated skeletal muscle cells can be identified by means of the provided markers of Table 8F, 8K and Table 9J. For example but not limited to, the differential methylation of HTF9C_HUMAN SEQ ID NO: 500 is used. According to Table 6, the differential methylation of HTF9C_HUMAN SEQ ID NO: 500 is a marker for skeletal muscle because the CpG dinucleotides of HTF9C_HUMAN SEQ ID NO: 500 are methylated within the range of 25-75% in skeletal muscle while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of HTF9C_HUMAN SEQ ID NO: 500 are, for example but not limited to, genomic DNA derived from or associated with HTF9C_HUMAN SEQ ID NO: 500; methylation specifically converted DNA derived from HTF9C_HUMAN SEQ ID NO: 500; mRNA, cDNA, protein, or peptide each of which derived at least in parts from HTF9C_HUMAN SEQ ID NO: 500. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for identifying fully in vitro differentiated muscle cell comprises:


1. Providing of a sample comprising genomic DNA of a skeletal muscle. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing the provided sample by Determining the methylation level of at least one CpG position within the sequence of HTF9C_HUMAN SEQ ID NO: 500 of the provided sample. Thereby a HTF9C_HUMAN profile is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Comparing the generated HTF9C_HUMAN profile with a HTF9C_HUMAN reference profile which is characteristic for a fully differentiated muscle cell. Wherein the determined HTF9C_HUMAN profile matches or is similar to the reference profile, the analyzed skeletal muscle cell is considered as fully differentiated.


In a preferred embodiment, the herein provided markers of Table 8F, 8K and Table 9J are used for diagnosing muscle cell associated diseases, in particular disease which are characterized by a death of muscle cells like muscular distrophy. The DNA of dead muscle cells is found in body fluids such as blood or urine. This DNA is identified by means of the herein provided markers of Table 8F, 8K and Table 9J. For example but not limited to, the differential methylation of EYA2 SEQ ID NO: 678 is used. According to Table 6, the differential methylation of EYA2 SEQ ID NO: 678 is a marker for skeletal muscle because the CpG dinucleotides of EYA2 SEQ ID NO: 678 are methylated within the range of 25-75% in skeletal muscle while other tissues show a different extend of methylation. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of EYA2 SEQ ID NO: 678 are, for example but not limited to, genomic DNA derived from or associated with EYA2 SEQ ID NO: 678; methylation specifically converted DNA derived from EYA2 SEQ ID NO: 678; mRNA, cDNA, protein, or peptide each of which derived at least in parts from EYA2 SEQ ID NO: 678. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for diagnosing muscle cell associated diseases comprises:


1. Providing of a sample derived from blood or urine, the sample comprising genomic DNA. The genomic DNA is isolated/purified from the provided sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Characterizing the sample by Determining the methylation level of at least one CpG position within the sequence of EYA2 SEQ ID NO: 678 of the provided sample. Thereby a EAY2 profile is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Deducing the presence or absence of a muscle associated disease from comparison of the determined EYA2 profile with at least one EYA2 reference profile. A EYA2 reference profile comprises the same position(s) as the determined profile. In addition a EYA2 profile is specific for either skeletal muscle cells (as herein provided by Table 8F, 8K and Table 9J), blood or urine comprising no skeletal muscle cell DNA, or blood or urine derived from a healthy individual.


CD8 T-Lymphocytes

In a preferred embodiment, the herein provided markers of Table 8A specific only for CD8 T-lymphocytes are used for quantifying CD8 T-lymphocytes, in particular for monitoring the immune system of individuals infected with HIV. The periodically determining of the number of CD8 T-lymphocytes for patients infected with HIV is a standard procedure in the art. It is necessary to decide whether and when a drug or treatment is necessary, whether a drug or treatment is still effective, and which drug or treatment can be selected. The said is necessary with respect to the HIV infection itself but also with respect to secondary infection. For example but not limited to, the differential methylation of RP4-695O20_B.9 SEQ ID NO: 706 is used. According to FIG. 1.296 and to Table 6, the differential methylation of RP4-695O20_B.9 SEQ ID NO: 706 is a marker for CD8 T-lymphocytes because the CpG dinucleotides of RP4-695O20_B.9 SEQ ID NO: 706 are methylated within the range of 75-100% in CD8 T-lymphocytes, sperm and liver while other tissues show a different extend of methylation. Furthermore CD8 T-lymphocytes can easily be distinguished from sperm or liver morphologically or by means of other markers. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of RP4-695O20_B.9 SEQ ID NO: 706 are, for example but not limited to, genomic DNA derived from or associated with RP4-695O20_B.9 SEQ ID NO: 706; methylation specifically converted DNA derived from RP4-695O20_B.9 SEQ ID NO: 706; mRNA, cDNA, protein, or peptide each of which derived at least in parts from RP4-695O20_B.9 SEQ ID NO: 706. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


Correspondingly, for example but not limited to, a method for quantifying CD8 T-lymphocytes comprises:


1. Providing a sample comprising genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega). Preferably said sample is a blood sample, plasma sample, or urine sample.


2. Determining the methylation level of at least one CpG position within the sequence of RP4-695O20_B.9 SEQ ID NO: 706 of the provided sample. Thereby a RP4-695O20_B.9 profile specific of said sample is generated. A person skilled in the art knows how to determine the methylation state or the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Deducing the number of CD8 T-lymphocytes from the comparison of the determined RP4-695O20_B.9 profile with one or more RP4-695O20_B.9 reference profiles. Said reference profiles are specific for a defined number of CD8 T-lymphocytes. In addition the reference profiles are obtained from the same sample type as the provided sample. For example but not limited to, blood is the sample type for the provided sample and the reference profiles. In a preferred embodiment, wherein the determined RP4-695O20_B.9 profile matches or is similar to a reference profile specific for a defined number of CD8 T-lymphocytes, the said number of CD8 T-lymphocytes is present in the analyzed sample. In a preferred embodiment, a calibration curve is prepared form the reference profiles. The number of CD8 T-lymphocytes is then deduced by comparing the determined RP4-695O20_B.9 profile with the calibration curve. Of course, other algorithms as the are known in the art are also preferred.


In addition, for example but not limited to it, a method for quantifying CD8 T-lymphocytes comprises:

    • 1. Providing of a sample, comprising genomic DNA;
    • 2. Contacting the genomic DNA or a derivative of it with at least one probe which is specific for at least one differentially methylated CpG position of the marker RP4-695O20_B.9 SEQ ID NO: 706. Said probe is selected from the group comprising antibody; 5-methylcytosine specific antibody (e.g. AbCam Cat. No. ab1884); affinity binding protein; protein binding specifically methylated or unmethylated DNA like MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof; nucleic acid; DNA, RNA, PNA or nucleic acid derivative specific for the methylated sequence. In addition, the probe is labeled directly or indirectly with a dye, protein, enzyme, metal, bead or chemical compound suitable for detection.
    • 3. Performing a detection reaction by means of the probe and/or the label. A person skilled in the art is aware of suitable detection reactions. For example, but not limited to, the detection reaction comprises Rabbit Peroxidase Anti-Peroxidase (PAP) Soluble Complex (Rockland Catalog#: P300-002); radioactive labeled probes; or probes fluorescently labeled like DNA probes coupled with Cy5 (Invitrogen). A person skilled in the art knows further suitable methods for detection.
    • 4. Quantifying the detection reaction in a manner so that the detected signal is indicative for the amount of probe or label and therewith for the number of dead liver cell. A person knows suitable methods for quantification.


Alternatively, for example but not limited to, a method for quantifying CD8 T-lymphocytes comprises:


1. Providing of a sample, the sample comprising one or more cells or genomic DNA. The genomic DNA is purified from said sample, preferably by means of a kit. A person skilled in the art is aware of suitable kits. For example, but not limited to it, the kit for purification of genomic DNA is the DNeasy Tissue Kit (Qiagen) or the Wizard Genomic DNA Purification kit (Promega).


2. Determining the methylation level of at least one CpG position within the sequence of RP4-695O20_B.9 SEQ ID NO: 706 of the provided sample. A person skilled in the art knows how to determine the methylation level of one or more CpG positions. For example, but not limited to, the methylation state or level is determined by means of at least one selected from the group comprising amplification method, PCR method, isothermal amplification method, NASBA method, LCR method, methylation specific amplification method, MSP (Methylation Specific PCR) method, nested MSP method, HeavyMethyl™ method, detection method, methylation specific detection method, bisulfite sequencing method, detection by means of DNA-arrays, detection by means of oligonucleotide microarrays, detection by means of CpG-island-microarrays, detection by means of restriction enzymes, detection by means of methylation sensitive restriction enzymes simultaneous methylation specific amplification and detection method, COBRA method, real-time PCR, HeavyMethyl™ real time PCR method, MSP MethyLight™ method, MethyLight™ method, MethyLight™ Algo™ method, QM method, Headloop MethyLight™ method, HeavyMethyl™ MethyLight™ method, HeavyMethyl™ Scorpion™ method, MSP Scorpion™ method, Headloop Scorpion™ method, methylation sensitive primer extension, Ms-SNuPE (Methylation-sensitive Single Nucleotide Primer Extension) method, and proteins binding specifically methylated or unmethylated DNA like the proteins MeCP2, MBD1, MBD2, MBD4, Kaiso or any domain thereof like but not limited to the CXXC-3 domain of the MBD1 protein or methylation-specific antibodies, e.g. anti-5-methylcytosine antibodies.


3. Quantifying the number of CD8 T-lymphocytes by comparing the determined one or more methylation levels of the sample with the respective herein provided one or more methylation levels of CD8 T-lymphocytes and with at least one methylation level representing the correspondent tissue, group of cells, or cell. Thereby said correspondent tissue, group of cells, or cell is derived from a healthy individual or is completely free of CD8 T-lymphocytes.


CD4+ Lymphocytes:

In a preferred embodiment, the herein provided markers of Table 8A specific only for CD4 T-lymphocytes are used for quantifying CD4 T-lymphocytes, in particular for monitoring the immune system of individuals infected with HIV. The periodically determining of the number of CD4 T-lymphocytes for patients infected with HIV is a standard procedure in the art. It is necessary to decide whether and when a drug or treatment is necessary, whether a drug or treatment is still effective, and which drug or treatment can be selected. The said is necessary with respect to the HIV infection itself but also with respect to secondary infection. For example but not limited to, the differential methylation of SEQ ID NO: 652 (no gene associated) is used. According to Table 6, the differential methylation of SEQ ID NO: 652 is a marker for CD4 T-lymphocytes because the CpG dinucleotides of SEQ ID NO: 652 are methylated within the range of 25-75% in CD4 T-lymphocytes while other tissues show a different extend of methylation. For example but not limited to, the quantification of CD4 T-lymphocytes is carried out as the above described methods for quantifying CD8 T-lymphocytes wherein the marker RP4-695O20_B.9 SEQ ID NO: 706 is substituted by SEQ ID NO: 652. Of course, corresponding markers can also be alternatively used. Thereby corresponding markers of SEQ ID NO: 652 are, for example but not limited to, genomic DNA derived from or associated with SEQ ID NO: 652; methylation specifically converted DNA derived from SEQ ID NO: 652; mRNA, cDNA, protein, or peptide each of which derived at least in parts from SEQ ID NO: 652. If the case may be, a person skilled in the art knows how to adjust the presented procedures.


All methods named in the above embodiments are known in the art. They are described for example in EP06075376.1 or in PCT/US2006/014667 both of which is hereby incorporated by reference.


While the present invention has been described with specificity in accordance with certain of its preferred embodiments, the following EXAMPLES and TABLES serve only to illustrate the invention and are not intended to limit the invention within the principles and scope of the broadest interpretations and equivalent configurations thereof.


Tables









TABLE 1







Overview of the genes or genomic regions according ot the present invention, with HUGO


ID and the SEQ ID NO.











SEQ ID NO:


Gene
HUGO ID
Genomic












FLOT1, flotillin 1, ENSG00000137312
FLOT1
413


C6orf25, chromosome 6 open reading frame 25,
C6orf25
414


ENSG00000096148


VARS, valyl-tRNA synthetase, ENSG00000096171
VARS
415


major histocompatibility complex, class II, DP beta 1,
HLA-DPB1
416


OTTHUMG00000031076, HLA-DPB1


HLA-DRB5, major histocompatibility complex, class II, DR
HLA-DRB5
417


beta 5, OTTHUMG00000031027


COL11A2, collagen, type XI, alpha 2,
COL11A2
418


OTTHUMG00000031036


PRAME, Melanoma antigen preferentially expressed in
PRAME
419


tumors (Preferentially expressed antigen of melanoma) (OPA-


interacting protein 4) (OIP4), ENSG00000185686


ZNRF3 protein (Fragment), ENSG00000183579, ZNRF3 zinc
ZNRF3
420


and ring finger 3 (ZNRF3)


AP000357.2 (Vega gene ID), Pseudogene
AP000357.2 (Vega
421



gene ID)


AP000357.3 (Vega gene ID), Pseudogene
AP000357.3 (Vega
422



gene ID)


solute carrier family 7 (cationic amino acid transporter, y+
SLC7A4
423


system), member 4, OTTHUMG00000030129, SLC7A4


Myosin-18B (Myosin XVIIIb), ENSG00000133454,
MYO18B
424


MYO18B


Q6ICL0_HUMAN (Predicted UniProt/TrEMBL ID),
Q6ICL0_HUMAN
425


hypothetical protein FLJ3257; ENSG00000184004
(Predicted



UniProt/TrEMBL



ID)


FBLN1; fibulin 1; ENSG00000077942
FBLN1
426


CYP2D6; cytochrome P450, family 2, subfamily D,
CYP2D6
427


polypeptide 6; ENSG00000100197


AC008132.9 (Vega gene ID); Pseudogene;
AC008132.9 (Vega
428


OTTHUMG00000030688
gene ID)


glycoprotein Ib (platelet), beta polypeptide,
GP1BB
429


OTTHUMT00000075045, GP1BB-001


no gene associated

430


AC006548.8 (Vega gene ID)
AC006548.8 (Vega
431



gene ID)


OTTHUMG00000030650, AC005399.2, putative processed
AC005399.2 (Vega
432


transcribed
gene ID)


topoisomerase (DNA) III beta, OTTHUMG00000030764,
TOP3B
433


TOP3B


no gene associated

434


KB-1269D1.3 (Vega gene ID); Pseudogene;
KB-1269D1.3 (Vega
435


OTTHUMG00000030694
gene ID)


GPR24; G protein-coupled receptor 24; ENSG00000128285
GPR24
436


GAL3ST1; galactose-3-O-sulfotransferase 1;
GAL3ST1
437


ENSG00000128242


Cat eye syndrome critical region protein 5 precursor,
CECR5
438


ENSG00000069998, CECR5 and (head to head),


OTTHUMG00000030478, AC006946.7


HORMAD2; HORMA domain containing 2;
HORMAD2
439


ENSG00000176635


OTTHUMG00000030922, RP3-438O4.2
RP3-438O4.2
440



(Vega_gene ID)


NP_997357.1 (RefSeq peptide ID); ENSG00000169668
NP_997357.1
441



(RefSeq peptide ID)


OTTHUMG00000030574, AP000357.3, novel pseudogene
AP000357.2 (Vega
442



gene ID)


LA16c-4G1.2 (Vega gene ID); Pseudogene;
LA16c-4G1.2 (Vega
443


OTTHUMG00000030832
gene ID)


KB-226F1.11 (Vega gene ID), embryonic marker,
KB-226F1.11 (Vega
444


OTTHUMG00000030123
gene ID)


OTTHUMG00000030780, CTA-373H7.4, novel pseudogene
CTA-373H7.4 (Vega
445



gene ID)


RP1-47A17.8 (Vega gene ID); OTTHUMG00000030878
RP1-47A17.8 (Vega
446



gene ID);


RP4-539M6.7 (Vega gene ID); Pseudogene;
RP4-539M6.7 (Vega
447


OTTHUMG00000030918
gene ID)


CSDC2; cold shock domain containing C2, RNA binding;
CSDC2
448


ENSG00000172346


Gamma-parvin, ENSG00000138964, PARVG
PARVG
449


OTTHUMG00000030167, CTA-243E7.3
CTA-243E7.3 (Vega
450



gene ID)


Oncostatin M precursor (OSM), ENSG00000099985, OSM
OSM
451


Oncostatin M precursor (OSM), ENSG00000099985, OSM
OSM
452


Myosin-18B (Myosin XVIIIb), ENSG00000133454,
MYO18B
453


MYO18B


Q6ICL0_HUMAN (Predicted UniProt/TrEMBL ID),
Q6ICL0_HUMAN
454


hypothetical protein FLJ3257; ENSG00000184004
(Predicted



UniProt/TrEMBL



ID)


OTTHUMG00000030140, CTA-299D3.6
CTA-299D3.6 (Vega
455



gene ID)


GALR3; galanin receptor 3; ENSG00000128310
GALR3
456


GALR3; galanin receptor 3; ENSG00000128310
GALR3
457


IL2RB; interleukin 2 receptor, beta; ENSG00000100385
IL2RB
458


CTA-343C1.3 (Vega gene ID); Putative Processed transcript;
CTA-343C1.3 (Vega
459


OTTHUMG00000030151
gene ID)


CTA-941F9.6 (Vega_gene ID)
CTA-941F9.6
460



(Vega_gene ID)


CTA-941F9.6 (Vega_gene ID)
CTA-941F9.6
461



(Vega_gene ID)


LL22NC03-121E8.1 (Vega gene ID); Novel Protein coding;
LL22NC03-121E8.1
462


OTTHUMG00000030676
(Vega gene ID)


Cytohesin-4, ENSG00000100055, PSCD4
PSCD4
463


RP4-754E20_A.4 (Vega gene ID); Putative Processed
RP4-754E20_A.4
464


transcript; OTTHUMG00000030716
(Vega gene ID)


PIB5PA; phosphatidylinositol (4,5) bisphosphate 5-
PIB5PA
465


phosphatase, A; ENSG00000185133; embryonic marker


no gene associated

466


PLA2G3; ENSG00000100078; phospholipase A2, group III
PLA2G3
467


PLA2G3; ENSG00000100078; phospholipase A2, group III
PLA2G3
468


DGCR2; DiGeorge syndrome critical region gene 2;
DGCR2
469


ENSG00000070413


TCN2; transcobalamin II; macrocytic anemia;
TCN2
470


ENSG00000185339


IGLL1; immunoglobulin lambda-like polypeptide 1;
IGLL1
471


ENSG00000128322


RP1-29C18.7 (Vega gene ID); Novel Processed transcript;
RP1-29C18.7 (Vega
472


OTTHUMG00000030424
gene ID)


IGLC1; immunoglobulin lambda constant 1 (Mcg marker);
IGLC1
473


ENSG00000100208


APOBEC3B; apolipoprotein B mRNA editing enzyme,
APOBEC3B
474


catalytic polypeptide-like 3B; ENSG00000179750


CRYBB1; crystallin, beta B1; ENSG00000100122
CRYBB1
475


CRYBA4; crystallin, beta A4; ENSG00000196431
CRYBA4
476


sushi domain containing 2, ENSG00000099994, SUSD2
SUSD2
477


sushi domain containing 2, ENSG00000099994, SUSD2
SUSD2
478


OTTHUMG00000030870, Putative Processed transcript,
CTA-503F6.1 (Vega
479


CTA-503F6.1
gene ID)


embryonic marker, OTTHUMG00000030800, KB-1323B2.3
KB-1323B2.3 (Vega
480



gene ID)


no gene associated

481


IGLV1-44; immunoglobulin lambda variable 1-44;
IGLV1-44
482


ENSG00000186751


IGLV1-44; immunoglobulin lambda variable 1-44;
IGLV1-44
483


ENSG00000186751


OTTHUMG00000030922, RP3-438O4.2
RP3-438O4.2
484



(Vega_gene ID)


OTTHUMG00000030922, RP3-438O4.2
RP3-438O4.2
485



(Vega_gene ID)


APOL4; apolipoprotein L, 4; ENSG00000100336
APOL4
486


OTTHUMG00000030852, RP4-756G23.1, novel processed
RP4-756G23.1
487


transcript
(Vega gene ID)


ENSG00000100399, Q96E60_HUMAN
Q96E60_HUMAN
488



(Predicted



UniProt/TrEMBL



ID)


Neutrophil cytosol factor 4 (NCF-4) (Neutrophil NADPH
NCF4
489


oxidase factor 4) (p40-phox) (p40phox)., ENSG00000100365,


NCF4


Neutrophil cytosol factor 4 (NCF-4) (Neutrophil NADPH
NCF4
490


oxidase factor 4) (p40-phox) (p40phox)., ENSG00000100365,


NCF4


Somatostatin receptor type 3 (SS3R) (SSR-28), D
SSTR3
491


ENSG00000183473, SSTR3


Somatostatin receptor type 3 (SS3R) (SSR-28), D
SSTR3
492


ENSG00000183473, SSTR3


Bcl-2 interacting killer (Apoptosis inducer NBK) (BP4)
BIK
493


(BIP1)., ENSG00000100290, BIK


GAS2-like protein 1 (Growth arrest-specific 2-like 1) (GAS2-
GAS2L1
494


related protein on chromosome 22) (GAR22 protein),


ENSG00000185340, GAS2L1


RP3-355C18.2 (Vega gene ID)
RP3-355C18.2
495



(Vega gene ID)


SOX10; SRY (sex determining region Y)-box 10;
SOX10
496


ENSG00000100146


Gamma-parvin ENSG00000138964
PARVG
497


Caspase recruitment domain protein 10 (CARD-containing
CARD10
498


MAGUK protein 3) (Carma 3). ENSG00000100065, CARD10


ENSG00000100101, NP_077289.1
NP_077289.1
499


HTF9C; HpaII tiny fragments locus 9C; ENSG00000099899
HTF9C_HUMAN
500



(UniProt/Swiss-Prot



ID)


Oncostatin M precursor (OSM), ENSG00000099985, OSM
OSM
501


CTA-407F11.4 (Vega gene ID); Novel Processed transcript;
CTA-407F11.4
502


OTTHUMG00000030804
(Vega gene ID)


Q6ICL0_HUMAN (Predicted UniProt/TrEMBL ID),
Q6ICL0_HUMAN
503


hypothetical protein FLJ3257; ENSG00000184004
(Predicted



UniProt/TrEMBL



ID)


CTA-989H11.2 (Vega gene ID); Putative Processed transcript;
CTA-989H11.2
504


OTTHUMG00000030141
(Vega gene ID)


transmembrane protease, serine 6
TMPRSS6
505


HMG2L1; high-mobility group protein 2-like 1;
HMG2L1
506


ENSG00000100281


NP_001017964.1 (RefSeq peptide ID); hypothetical protein
NP_001017964.1
507


LOC150223; ENSG00000161179
(RefSeq peptide ID)


Platelet-derived growth factor B chain precursor (PDGF B-
PDGFB
508


chain), ENSG00000100311, PDGFB


OTTHUMG00000030815, CTA-384D8.15
CTA-384D8.15
509



(Vega gene ID)


MGAT3; mannosyl (beta-1,4-)-glycoprotein beta-1,4-N-
MGAT3
510


acetylglucosaminyltransferase; ENSG00000128268


Ceramide kinase (EC 2.7.1.138), Acylsphingosine kinase,
CERK
511


CERK, Lipid kinase 4, LK4, ENSG00000100422, CERK


Reticulon 4 receptor precursor (Nogo receptor) (NgR) (Nogo-
RTN4R
512


66 receptor), ENSG00000040608, RTN4R


UNC84B; unc-84 homolog B (C. Elegans);
UNC84B
513


ENSG00000100242


RABL4; RAB, member of RAS oncogene family-like 4;
RABL4
514


ENSG00000100360


Cadherin EGF LAG seven-pass G-type receptor 1 precursor
CELSR1
515


(Flamingo homolog 2) (hFmi2), ENSG00000075275,


CELSR1


OTTHUMG00000030326, LL22NC03-5H6.1
LL22NC03-5H6.1
516



(Vega gene ID)


OTTHUMG00000030656, RP3-515N1.6
RP3-515N1.6 (Vega
517



gene ID)


SMTN; smoothelin; ENSG00000183963
SMTN
518


ZNRF3 protein (Fragment), ENSG00000183579, ZNRF3 zinc
ZNRF3
519


and ring finger 3 (ZNRF3)


OTTHUMG00000030700, GRB2-related adaptor protein 2,
GRAP2
520


GRAP2


CAP-binding protein complex interacting protein 1 isoform a
NP_073622.2
521


Source: RefSeq_peptide NP_073622
(RefSeq peptide ID)


SAM50_HUMAN (UniProt/Swiss-Prot ID),
SAMM50
522


ENSG00000100347, SAM50-like protein CGI-51; sorting


and assembly machinery component 50 homolog (S. Cerevisiae)


SULT4A1; sulfotransferase family 4A, member 1;
SULT4A1
523


ENSG00000130540


TIMP3; TIMP metallopeptidase inhibitor 3 (Sorsby fundus
TIMP3
524


dystrophy, pseudoinflammatory); ENSG00000100234


T-box transcription factor TBX1 (T-box protein 1) (Testis-
TBX1
525


specific T-box protein), ENSG00000184058, TBX1, TBX1


is involved in heart development-Great


ENSG00000186732, metallophosphoesterase domain
MPPED1
526


containing 1, NM_001585.2


ENSG00000188511, NP_942148.1 novel Gene hypothetical
NP_942148.1
527


protein LOC348645
(RefSeq peptide ID)


Cdc42 effector protein 1, ENSG00000128283, CDC42EP1
CDC42EP1
528


RPL3; ribosomal protein L3; ENSG00000100316
RPL3
529


APOL2; apolipoprotein L, 2; ENSG00000128335
APOL2
530


RAC2; ras-related C3 botulinum toxin substrate 2 (rho family,
RAC2
531


small GTP binding protein Rac2); ENSG00000128340


OTTHUMP00000028917, ENSG00000100399,
Q96E60_HUMAN
532


Q96E60_HUMAN
(Predicted



UniProt/TrEMBL



ID)


Neutrophil cytosol factor 4 (NCF-4) (Neutrophil NADPH
NCF4
533


oxidase factor 4) (p40-phox) (p40phox)., ENSG00000100365,


NCF4


XP_371837.1 (RefSeq peptide predicted ID); PREDICTED:
XP_371837.1
534


similar to oxidoreductase UCPA Source:
(RefSeq peptide


RefSeq_peptide_predicted XP_371837; ENSG00000168768
predicted ID)


triggering receptor expressed on myeloid cells-like 2,
TREML2
535


ENSG00000112195, TREML2


TREML1; triggering receptor expressed on myeloid cells-like
TREML1
536


1; ENSG00000161911


ENSG00000178199, Q6ZRW2_HUMAN; zinc finger
ZC3H12D
537


CCCH-type containing 12D


AIM1; absent in melanoma1; ENSG00000112297
AIM1
538


NKG2D ligand 4 precursor (NKG2D ligand 4) (NKG2DL4)
RAET1E
539


(N2DL-4) (Retinoic acid early transcript 1E) (Lymphocyte


effector toxicity activation ligand) (RAE-1-like transcript 4)


(RL-4), ENSG00000164520, RAET1E


Disheveled associated activator of morphogenesis 2,
DAAM2
540


ENSG00000146122, DAAM2


RP11-535K1.1 (Vega gene ID); Putative Processed transcript;
RP11-535K1.1
541


OTTHUMG00000014660
(Vega gene ID)


OTTHUMG00000015679; Novel Protein coding; RP3-
RP3-509I19.3 (Vega
542


509I19.3
gene ID)


RP11-503C24.1 (Vega gene ID); Putative Processed
RP11-503C24.1
543


transcript; OTTHUMG00000016040
(Vega gene ID)


GABRR2; gamma-aminobutyric acid (GABA) receptor, rho
GABRR2
544


2; ENSG00000111886


ANKRD6; ankyrin repeat domain 6; ENSG00000135299
ANKRD6
545


TXLNB; taxilin beta; ENSG00000164440
TXLNB
546


TXLNB; taxilin beta; ENSG00000164440
TXLNB
547


RP5-899B16.2 (Vega gene ID); Putative Processed transcript;
RP5-899B16.2
548


OTTHUMG00000015698
(Vega gene ID)


Probable G-protein coupled receptor 116 precursor,
GPR116
549


ENSG00000069122, GPR116


RP11-146I2.1 (Vega gene ID); Novel Processed transcript;
RP11-146I2.1 (Vega
550


OTTHUMG00000014290
gene ID)


GPR115; G protein-coupled receptor 115;
GPR115
551


ENSG00000153294


GPR126; G protein-coupled receptor 126;
GPR126
552


ENSG00000112414 embryonic marker


RP1-60O19.1 (Vega gene ID); Known Processed transcript;
RP1-60O19.1 (Vega
553


OTTHUMG00000015305
gene ID)


OTTHUMG00000015313, RP1-47M23.1 SCML4 sex comb
SCML4
554


on midleg-like 4 (Drosophila) [Homo sapiens]


OTTHUMG00006004170, TPX1testis specific protein 1
CRISP2
555


(probe H4-1 p3-1)


OTTHUMG00000014829, RP11-397G17.1, novel processed
RP11-397G17.1
556


transcript.
(Vega gene ID)


OTTHUMG00000015337RP11-487F23.3 hypothetical
RP11-487F23.3
557


LOC389422
(Vega gene ID)


Nesprin-1 (Nuclear envelope spectrin repeat protein 1)
SYNE1
558


(Synaptic nuclear envelope protein 1) (Syne-1) (Myocyte


nuclear envelope protein 1) (Myne-1) (Enaptin),


ENSG00000131018, SYNE1


Nesprin-1 (Nuclear envelope spectrin repeat protein 1)
SYNE1
559


(Synaptic nuclear envelope protein 1) (Syne-1) (Myocyte


nuclear envelope protein 1) (Myne-1) (Enaptin),


ENSG00000131018, SYNE1


RP11-398K22.4 (Vega gene ID); Putative Processed
RP11-398K22.4
560


transcript; OTTHUMG00000015024
(Vega gene ID)


MyoD family inhibitor (Myogenic repressor I-mf),
MDFI
561


ENSG00000112559, MDFI


OTTHUMG00000014691, putative processed transcript,
RP11-533O20.2
562


RP11-533O20.2
(Vega gene ID)


RP3-398D13.4 (Vega gene ID); OTTHUMG00000014188
RP3-398D13.4
563



(Vega gene ID);


RP3-429O6.1 (Vega gene ID); Putative Processed transcript;
RP3-429O6.1 (Vega
564


OTTHUMG00000014195
gene ID)


MOG; myelin oligodendrocyte glycoprotein;
MOG
565


ENSG00000137345


RP3-495K2.2 (Vega gene ID); Putative Processed transcript;
RP3-495K2.2 (Vega
566


OTTHUMG00000016052
gene ID)


RP11-417E7.1 (Vega gene ID); Putative Processed transcript;
RP11-417E7.1 (Vega
567


OTTHUMG00000016054
gene ID)


yrosine-protein kinase-like 7 precursor (Colon carcinoma
PTK7
568


kinase 4) (CCK-4)., ENSG00000112655, PTK7


RP11-174C7.4 (Vega gene ID)
RP11-174C7.4
569



(Vega gene ID)


cytidine monophosphate-N-acetylneuraminic acid
CMAH
570


hydroxylase (CMP-N-acetylneuraminate monooxygenase);


CMAH


PKHD1; polycystic kidney and hepatic disease 1 (autosomal
PKHD1
571


recessive); ENSG00000170927


RP3-471C18.2 (Vega gene ID); Novel Processed transcript;
RP3-471C18.2
572


OTTHUMG00000014332
(Vega gene ID)


RP11-204E9.1 (Vega gene ID); Putative Processed transcript;
RP11-204E9.1 (Vega
573


OTTHUMG00000014342
gene ID)


glutathione peroxidase 5, OTTHUMG00000016307, GPX5
GPX5
574


RP11-411K7.1 (Vega gene ID); Putative Processed transcript;
RP11-411K7.1
575


OTTHUMG00000014887
(Vega gene ID)


skin marker, Glutamate receptor, ionotropic kainate 2
GRIK2
576


precursor (Glutamate receptor 6) (GluR-6) (GluR6)


(Excitatory amino acid receptor 4) (EAA4)


C6orf142; chromosome 6 open reading frame 142;
C6orf142
577


ENSG00000146147


HDGFL1; hepatoma derived growth factor-like 1;
HDGFL1
578


ENSG00000112273


forkhead box C1, OTTHUMG00000016182, FOXC1
FOXC1
579


C6orf188; chromosome 6 open reading frame 188;
C6orf188
580


ENSG00000178033


ME1; malic enzyme 1, NADP(+)-dependent, cytosolic;
ME1
581


ENSG00000065833


SLC22A1; solute carrier family 22 (organic cation
SLC22A1
582


transporter), member 1


RP11-235G24.1 (Vega gene ID)
RP11-235G24.1
583



(Vega gene ID)


T-box 18; TBX18
TBX18
584


CTA-31J9.2, putative processed transcript,
CTA-31J9.2 (Vega
585


OTTHUMG00000015619
gene ID)


RP1-32B1.4 (Vega gene ID); Putative Processed transcript
RP1-32B1.4 (Vega
586


OTTHUMG00000015628
gene ID)


OTTHUMG00000014223, RP11-203H2.2, novel processed
RP11-203H2.2
587


treanscript
(Vega gene ID)


OTTHUMG00000014737, C6orf154 and Name: chromosome
C6orf154
588


6 open reading frame 154; RP3-337H4.2


transcription factor AP-2 alpha, OTTHUMG00000014235,
TFAP2A
589


TFAP2A


IL20RA; interleukin 20 receptor, alpha; ENSG00000016402
IL20RA
590


KAAG1; kidney associated antigen 1; ENSG00000146049
KAAG1
591


TGM3; transglutaminase 3 (E polypeptide, protein-glutamine-
TGM3
592


gamma-glutamyltransferase); ENSG00000125780


RASSF2; Ras association (RalGDS/AF-6) domain family 2;
RASSF2
593


ENSG00000101265


no gene associated

594


no gene associated

595


no gene associated

596


no gene associated

597


no gene associated

598


no gene associated

599


no gene associated

600


no gene associated

601


no gene associated

602


no gene associated

603


no gene associated

604


RP4-697P8.2 (Vega gene ID); Putative Processed transcript;
RP4-697P8.2 (Vega
605


OTTHUMG00000031879
gene ID)


no gene associated

606


OTTHUMG00000031883,
RP4-734C18.1
607


RP4-734C18.1, putative processed transcript
(Vega gene ID)


no gene associated

608


no gene associated

609


no gene associated

610


no gene associated

611


no gene associated

612


Ras and Rab interactor 2,
RIN2
613


OTTHUMG00000031996, RIN2


no gene associated

614


no gene associated

615


no gene associated

616


no gene associated

617


no gene associated

618


no gene associated

619


no gene associated

620


no gene associated

621


no gene associated

622


no gene associated

623


C20orf112; chromosome 20 open reading frame 112;
C20orf112
624


OTTHUMG00000032219


FER1L4; fer-1-like 4 (C. Elegans); OTTHUMG00000032354
FER1L4
625


no gene associated

626


no gene associated

627


Protein C20orf102 precursor, ENSG00000132821,

628


CT102_HUMAN


no gene associated

629


no gene associated

630


no gene associated

631


no gene associated

632


no gene associated - Nearest transcript CDH22 (~18 kb

633


upstream)


no gene associated

634


no gene associated

635


no gene associated

636


no gene associated

637


no gene associated

638


no gene associated

639


no gene associated

640


ZHX3; zinc fingers and homeoboxes 3;
ZHX3
641


OTTHUMG00000032481


no gene associated

642


CHD6; chromodomain helicase DNA binding protein 6;
CHD6
643


ENSG00000124177


no gene associated

644


PTPRG; protein tyrosine phosphatase, receptor type G;
PTPRG
645


ENSG00000144724


no gene associated

646


no gene associated

647


no gene associated

648


PTPNS1; protein tyrosine phosphatase, non-receptor type
PTPNS1
649


substrate 1; ENSG00000198053


Q7Z5T1_HUMAN (Predicted UniProt/TrEMBL ID);
Q7Z5T1_HUMAN
650


KIAA1442 protein; ENSG00000088881
(Predicted



UniProt/TrEMBL



ID)


NP_689717.2 (RefSeq peptide ID); ENSG00000171984
NP_689717.2
651



(RefSeq peptide ID)


ENSG00000149346, NP_001009608.1, hypothetical protein
C20orf94
652


LOC128710, chromosome 20 open reading frame 94


C20orf82; chromosome 20 open reading frame 82;
C20orf82
653


ENSG00000101230


C20orf23; chromosome 20 open reading frame 23;
C20orf23
654


ENSG00000089177; embryonic marker


PCSK2; proprotein convertase subtilisin/kexin type 2;
PCSK2
655


ENSG00000125851


PCSK2; proprotein convertase subtilisin/kexin type 2;
PCSK2
656


ENSG00000125851


solute carrier family 24 (sodiumVpotassiumVcalcium
SLC24A3
657


exchanger), member 3, OTTHUMG00000031993,


SLC24A3


solute carrier family 24 (sodiumVpotassiumVcalcium
SLC24A3
658


exchanger), member 3, OTTHUMG00000031993,


SLC24A3


ENSG00000089101, CT026_HUMAN
C20orf26
659


ENSG00000089101, CT026_HUMAN
C20orf26
660


C20orf74 protein, ENSG00000188559, Q9ULE8_HUMAN
Q9ULE8_HUMAN
661



(Predicted



UniProt/TrEMBL



ID)


C20orf74 protein, ENSG00000188559, Q9ULE8_HUMAN
Q9ULE8_HUMAN
662



(Predicted



UniProt/TrEMBL



ID)


C20orf74 protein, ENSG00000188559, Q9ULE8_HUMAN
Q9ULE8_HUMAN
663



(Predicted



UniProt/TrEMBL



ID)


PLAGL2; pleiomorphic adenoma gene-like 2;
PLAGL2
664


ENSG00000126003


GGTL3; gamma-glutamyltransferase-like 3;
GGTL3
665


ENSG00000131067


MYH7B; myosin, heavy polypeptide 7B, cardiac muscle,
MYH7B
666


beta; ENSG00000078814


TRPC4AP; transient receptor potential cation channel,
TRPC4AP
667


subfamily C, member 4 associated protein;


ENSG00000100991


EPB41L1; erythrocyte membrane protein band 4.1-like 1;
EPB41L1
668


ENSG00000088367


C20orf117; chromosome 20 open reading frame 117;
C20orf117
669


OTTHUMG00000032395


PTPRT; protein tyrosine phosphatase, receptor type, T;
PTPRT
670


ENSG00000196090


PTPRT; protein tyrosine phosphatase, receptor type, T;
PTPRT
671


ENSG00000196090


PTPRT; protein tyrosine phosphatase, receptor type, T;
PTPRT
672


ENSG00000196090


PTPRT; protein tyrosine phosphatase, receptor type, T;
PTPRT
673


ENSG00000196090


PTPRT; protein tyrosine phosphatase, receptor type, T;
PTPRT
674


ENSG00000196090


SDC4; syndecan 4 (amphiglycan, ryudocan);
SDC4
675


ENSG00000124145


SDC4; syndecan 4 (amphiglycan, ryudocan);
SDC4
676


ENSG00000124145


cadherin-like 22, OTTHUMG00000033073, CDH22
CDH22
677


EYA2; eyes absent homolog 2 (Drosophila);
EYA2
678


ENSG00000064655


SULF2; sulfatase 2; ENSG00000196562
SULF2
679


KCNB1; potassium voltage-gated channel, Shab-related
KCNB1
680


subfamily, member 1; ENSG00000158445


Breast carcinoma amplified sequence 4,
BCAS4
681


ENSG00000124243,


BCAS4


nuclear factor of activated T-cells, cytoplasmic, calcineurin-
NFATC2
682


dependent 2, OTTHUMG00000032747, NFATC2


Nuclear factor of activated T-cells, cytoplasmic 2 (T cell
NFATC2
683


transcription factor NFAT1) (NFAT pre-existing subunit)


(NF-ATp), ENSG00000101096, NFATC2


Bone morphogenetic protein 7 precursor (BMP-7)
BMP7
684


(Osteogenic protein 1) (OP-1) (Eptotermin alfa),


ENSG00000101144, BMP7


transmembrane, prostate androgen induced RNA,
TMEPAI
685


OTTHUMG00000032831, TMEPAI


ENSG00000176659, NP_775915.1
NP_775915.1
686



(RefSeq peptide ID)


CDH4; cadherin 4, type 1, R-cadherin (retinal);
CDH4
687


ENSG00000179242


NP_001002034.1 (RefSeq peptide ID); ENSG00000177096
NP_001002034.1
688



(RefSeq peptide ID)


NP_612444.1 (RefSeq peptide ID); ENSG00000133477
NP_612444.1
689



(RefSeq peptide ID)


no gene associated

690


OTTHUMG00000030780, CTA-373H7.4, novel pseudogene
CTA-373H7.4 (Vega
691



gene ID)


no gene associated

692


Cat eye syndrome critical region protein 1 precursor,
CECR1
693


ENSG00000093072, CECR1


IGLC1; immunoglobulin lambda constant 1 (Mcg marker);
IGLC1
694


ENSG00000100208


OTTHUMG00000030521, AC000095.4 putative processed
AC000095.4 (Vega
695


transcript;
gene ID)


Uroplakin-3A precursor (Uroplakin III) (UPIII).,
UPK3A
696


ENSG00000100373, UPK3A


Sp1 site_no gene associated

697


USP18; ubiquitin specific peptidase 18;
USP18
698


OTTHUMG00000030949


BCR; breakpoint cluster region; ENSG00000186716
BCR
699


TBC1D10A; TBC1 domain family, member 10A;
TBC1D10A
700


ENSG00000099992


signal peptide-CUB domian-EGF-related 1,
SCUBE1
701


ENSG00000159307, SCUBE1


MAPK8IP2; mitogen-activated protein kinase 8 interacting
MAPK8IP2
702


protein 2; ENSG00000008735


ENSG00000192797, miRNA
Not Available
703


RPL3; ribosomal protein L3; ENSG00000100316
RPL3
704


RPL3; ribosomal protein L3; ENSG00000100316
RPL3
705


RP4-695O20_B.9 (Vega gene ID); Putative Processed
RP4-695O20_B.9
706


transcript; OTTHUMG00000030111
(Vega gene ID)


No associated gene

707


MN1; meningioma (disrupted in balanced translocation) 1;
MN1
708


ENSG00000169184


no gene associated

709


RTDR1; rhabdoid tumor deletion region gene 1;
RTDR1
710


ENSG00000100218


RPL3; ribosomal protein L3; ENSG00000100316
RPL3
711


embryonic marker, GRB2-related adaptor protein 2,
GRAP2
712


OTTHUMG00000030700, GRAP2


Serine/threonine-protein kinase 19 (EC 2.7.1.37) (RP1
STK19
713


protein) (G11 protein).


Transcription factor 19 (Transcription factor SC1).
TCF19_HUMAN
714


Pannexin-2
PANX2
715



CTA-243E7.3
716


signal peptide-CUB domian-EGF-related 1
SCUBE1
717


Reticulon 4 receptor precursor (Nogo receptor) (NgR) (Nogo-
RTN4R
718


66 receptor)


Arylsulfatase A precursor (EC 3.1.6.8) (ASA) (Cerebroside-
ARSA
719


sulfatase) [Contains: Arylsulfatase A component B;


Arylsulfatase A component C]


glycoprotein Ib (platelet), beta polypeptide
GP1BB
720



No gene associated
721



No gene associated
722


Mitochondrial glutamate carrier 2 (Glutamate/H(+) symporter
SLC25A18
723


2) (Solute carrier family 25 member 18, ENSG00000182902,


SLC25A18


Thioredoxin reductase 2, mitochondrial precursor (EC
TXNRD2
724


1.8.1.9) (TR3) (TR-beta) (Selenoprotein Z) (SelZ)


Somatostatin receptor type 3 (SS3R) (SSR-28)
SSTR3
725



RP11-191L9.1
726


No description-pseudogene
AP000357.3
727


Cat eye syndrome critical region protein 1 precursor
CECR1
728



No gene associated
729


Membrane protein MLC1
MLC1
730


BAI1-associated protein 2-like 2
BAIAP2L2
731


No description
NP_056185.1
732


No description
RP4-695O20_B.9
733


OTTHUMG00000030167, CTA-243E7.3
CTA-243E7.3 (Vega
734



gene ID)


novel transcript
XXbac-B444P24.7
735



LL22NC03-121E8.1-
736



001


No description
Q6ZN90_HUMAN
737


NFAT activation molecule 1 precursor (Calcineurin/NFAT-
NFAM1
738


activating ITAM-containing protein) (NFAT activating


protein with ITAM motif 1).


immunoglobulin lambda constant 2
IGLC2
739


immunoglobulin lambda constant 2
IGLC2
740


OTTHUMG00000030870, CTA-503F6.1
CTA-503F6.1
741



(Vega_gene ID)


Lactosylceramide 4-alpha-galactosyltransferase (EC
A4GALT
742


2.4.1.228)



RP11-191L9.3
743


Cold shock domain protein C2 (RNA-binding protein PIPPin)
CSDC2_HUMAN
744


GAS2-like protein 1 (Growth arrest-specific 2-like 1) (GAS2-
GAS2L1
745


related protein on chromosome 22) (GAR22 protein),


ENSG00000185340, GAS2L1


BAI1-associated protein 2-like 2
BAIAP2L2
746



NP_997360.1
747


OTTHUMG00000030991, LL22NC03-75B3.6
LL22NC03-75B3.6
748



(Vega gene ID)


Reticulon 4 receptor precursor (Nogo receptor) (NgR) (Nogo-
RTN4R
749


66 receptor)


Smoothelin
SMTN
750


solute carrier family 35, member E4
SLC35E4
751


Protein C22orf13 (Protein LLN4)
CV013_HUMAN
752



No gene associated
753


Histone
HIST1H3A
754


Gamma-aminobutyric-acid receptor rho-1 subunit precursor
GABRR1
755


(GABA(A) receptor).


OTTHUMG00000015693, RP11-12A2.3
RP11-12A2.3
756



(Vega_gene ID)



RP5-899B16.1
757



RP11-146I2.2
758



NP_060483.2
759


Forkhead box protein O3A, ENSG00000118689, FOXO3A
FOXO3A
760


nuclear receptor coactivator 7
NCOA7
761



RP11-554D15.1
762


chromosome 6 open reading frame 190
C6orf190
763


phosphatase and actin regulator 2
PHACTR2
764


High mobility group protein HMG-I/HMG-Y, HMG-I(Y),
HMGA1
765


High mobility group AT-hook 1, High mobility group protein


A1, ENSG00000137309, HMGA1


Pantetheinase precursor (EC 3.5.1.—), ENSG00000112299,
VNN1
766


VNN1


histone H2A
HIST1H2AA
767


transcription factor AP-2 alpha (activating enhancer binding
TFAP2A
768


protein 2 alpha)


N-acetyllactosaminide beta-1,6-N-acetylglucosaminyl-
GCNT2
769


transferase (EC 2.4.1.150), ENSG00000111846, GCNT2



No gene associated
770



No gene associated
771



No gene associated
772



No gene associated
773



No gene associated
774



No gene associated
775



No gene associated
776



No gene associated
777



No gene associated
778



No gene associated
779



RP11-318C17.1
780



No gene associated
781



No gene associated
782



No gene associated
783



No gene associated
784



No gene associated
785



No gene associated
786


novel transcript
RP11-216C10.1
787



No gene associated
788



No gene associated
789



No gene associated
790



RP11-410N8.3
791


TIMP3; TIMP metallopeptidase inhibitor 3 (Sorsby fundus
TIMP3
792


dystrophy, pseudoinflammatory); ENSG00000100234



No gene associated
793



No gene associated
794



No gene associated
795



No gene associated
796



No gene associated
797



No gene associated
798



No gene associated
799



No gene associated
800



No gene associated
801



No gene associated
802



No gene associated
803


sorting nexin 5
SNX5
804


Probable D-tyrosyl-tRNA(Tyr) deacylase (EC 3.1.—.—)
HARS2
805


solute carrier family 24 (sodium/potassium/calcium
SLC24A3
806


exchanger) member 3, OTTHUMG00000031993, SLC24A3


ENSG00000089101, CT026_HUMAN, C20orf26
CT026_HUMAN
807


RNA-binding protein Raly (hnRNP associated with lethal
RALY
808


yellow homolog) D; ENSG00000125970, RALY


Protein phosphatase 1 regulatory inhibitor subunit 16B (TGF-
PPP1R16B
809


beta-inhibited membrane-associated protein) (hTIMAP)


(CAAX box protein TIMAP) (Ankyrin repeat domain protein


4)


protein tyrosine phosphatase, receptor type, T
PTPRT
810


protein tyrosine phosphatase, receptor type, T
PTPRT
811


protein tyrosine phosphatase, receptor type, T
PTPRT
812


Receptor-type tyrosine-protein phosphatase T precursor (EC
PTPRT
813


3.1.3.48) (R-PTP-T) (RPTP-rho)


cadherin-like 22
CDH22
814


potassium voltage-gated channel, Shab-related subfamily,
KCNB1
815


member 1


potassium voltage-gated channel, Shab-related subfamily,
KCNB1
816


member 1


Zinc finger protein SNAI1 (Snail protein homolog) (Sna
SNAI1
817


protein)


Cadherin-4 precursor (Retinal-cadherin) (R-cadherin) (R-
CDH4
818


CAD)


cadherin 4, type 1, R-cadherin (retinal)
CDH4
819


Cadherin-4 precursor (Retinal-cadherin) (R-cadherin) (R-
CDH4
820


CAD)


Metalloproteinase inhibitor 3 precursor (TIMP-3) (Tissue
TIMP3
821


inhibitor of metalloproteinases-3) (MIG-5 protein).


Tubulin alpha-8 chain (Alpha-tubulin 8)
TUBA8
822



No gene associated
823



No gene associated
824
















TABLE 2







Overview of SEQ ID NO of the genes or genomic regions and the


SEQ ID NOs of corresponding bisulfite treated nucleic acids (DNA


upmethylated sense strand, DNA upmethylated antisense strand,


DNA downmethylated sense strand, DNA downmethylated antisense


strand).














SEQ ID NO:
SEQ ID NO:


SEQ ID
SEQ ID NO
SEQ ID NO:
Down-
Down-


NO:
Upmethylated
Upmethylated
methylated
methylated


Genomic
Sense
Antisense
Sense
Antisense





413
1649
1650
3297
3298


414
1651
1652
3299
3300


415
1653
1654
3301
3302


416
1655
1656
3303
3304


417
1657
1658
3305
3306


418
1659
1660
3307
3308


419
1661
1662
3309
3310


420
1663
1664
3311
3312


421
1665
1666
3313
3314


422
1667
1668
3315
3316


423
1669
1670
3317
3318


424
1671
1672
3319
3320


425
1673
1674
3321
3322


426
1675
1676
3323
3324


427
1677
1678
3325
3326


428
1679
1680
3327
3328


429
1681
1682
3329
3330


430
1683
1684
3331
3332


431
1685
1686
3333
3334


432
1687
1688
3335
3336


433
1689
1690
3337
3338


434
1691
1692
3339
3340


435
1693
1694
3341
3342


436
1695
1696
3343
3344


437
1697
1698
3345
3346


438
1699
1700
3347
3348


439
1701
1702
3349
3350


440
1703
1704
3351
3352


441
1705
1706
3353
3354


442
1707
1708
3355
3356


443
1709
1710
3357
3358


444
1711
1712
3359
3360


445
1713
1714
3361
3362


446
1715
1716
3363
3364


447
1717
1718
3365
3366


448
1719
1720
3367
3368


449
1721
1722
3369
3370


450
1723
1724
3371
3372


451
1725
1726
3373
3374


452
1727
1728
3375
3376


453
1729
1730
3377
3378


454
1731
1732
3379
3380


455
1733
1734
3381
3382


456
1735
1736
3383
3384


457
1737
1738
3385
3386


458
1739
1740
3387
3388


459
1741
1742
3389
3390


460
1743
1744
3391
3392


461
1745
1746
3393
3394


462
1747
1748
3395
3396


463
1749
1750
3397
3398


464
1751
1752
3399
3400


465
1753
1754
3401
3402


466
1755
1756
3403
3404


467
1757
1758
3405
3406


468
1759
1760
3407
3408


469
1761
1762
3409
3410


470
1763
1764
3411
3412


471
1765
1766
3413
3414


472
1767
1768
3415
3416


473
1769
1770
3417
3418


474
1771
1772
3419
3420


475
1773
1774
3421
3422


476
1775
1776
3423
3424


477
1777
1778
3425
3426


478
1779
1780
3427
3428


479
1781
1782
3429
3430


480
1783
1784
3431
3432


481
1785
1786
3433
3434


482
1787
1788
3435
3436


483
1789
1790
3437
3438


484
1791
1792
3439
3440


485
1793
1794
3441
3442


486
1795
1796
3443
3444


487
1797
1798
3445
3446


488
1799
1800
3447
3448


489
1801
1802
3449
3450


490
1803
1804
3451
3452


491
1805
1806
3453
3454


492
1807
1808
3455
3456


493
1809
1810
3457
3458


494
1811
1812
3459
3460


495
1813
1814
3461
3462


496
1815
1816
3463
3464


497
1817
1818
3465
3466


498
1819
1820
3467
3468


499
1821
1822
3469
3470


500
1823
1824
3471
3472


501
1825
1826
3473
3474


502
1827
1828
3475
3476


503
1829
1830
3477
3478


504
1831
1832
3479
3480


505
1833
1834
3481
3482


506
1835
1836
3483
3484


507
1837
1838
3485
3486


508
1839
1840
3487
3488


509
1841
1842
3489
3490


510
1843
1844
3491
3492


511
1845
1846
3493
3494


512
1847
1848
3495
3496


513
1849
1850
3497
3498


514
1851
1852
3499
3500


515
1853
1854
3501
3502


516
1855
1856
3503
3504


517
1857
1858
3505
3506


518
1859
1860
3507
3508


519
1861
1862
3509
3510


520
1863
1864
3511
3512


521
1865
1866
3513
3514


522
1867
1868
3515
3516


523
1869
1870
3517
3518


524
1871
1872
3519
3520


525
1873
1874
3521
3522


526
1875
1876
3523
3524


527
1877
1878
3525
3526


528
1879
1880
3527
3528


529
1881
1882
3529
3530


530
1883
1884
3531
3532


531
1885
1886
3533
3534


532
1887
1888
3535
3536


533
1889
1890
3537
3538


534
1891
1892
3539
3540


535
1893
1894
3541
3542


536
1895
1896
3543
3544


537
1897
1898
3545
3546


538
1899
1900
3547
3548


539
1901
1902
3549
3550


540
1903
1904
3551
3552


541
1905
1906
3553
3554


542
1907
1908
3555
3556


543
1909
1910
3557
3558


544
1911
1912
3559
3560


545
1913
1914
3561
3562


546
1915
1916
3563
3564


547
1917
1918
3565
3566


548
1919
1920
3567
3568


549
1921
1922
3569
3570


550
1923
1924
3571
3572


551
1925
1926
3573
3574


552
1927
1928
3575
3576


553
1929
1930
3577
3578


554
1931
1932
3579
3580


555
1933
1934
3581
3582


556
1935
1936
3583
3584


557
1937
1938
3585
3586


558
1939
1940
3587
3588


559
1941
1942
3589
3590


560
1943
1944
3591
3592


561
1945
1946
3593
3594


562
1947
1948
3595
3596


563
1949
1950
3597
3598


564
1951
1952
3599
3600


565
1953
1954
3601
3602


566
1955
1956
3603
3604


567
1957
1958
3605
3606


568
1959
1960
3607
3608


569
1961
1962
3609
3610


570
1963
1964
3611
3612


571
1965
1966
3613
3614


572
1967
1968
3615
3616


573
1969
1970
3617
3618


574
1971
1972
3619
3620


575
1973
1974
3621
3622


576
1975
1976
3623
3624


577
1977
1978
3625
3626


578
1979
1980
3627
3628


579
1981
1982
3629
3630


580
1983
1984
3631
3632


581
1985
1986
3633
3634


582
1987
1988
3635
3636


583
1989
1990
3637
3638


584
1991
1992
3639
3640


585
1993
1994
3641
3642


586
1995
1996
3643
3644


587
1997
1998
3645
3646


588
1999
2000
3647
3648


589
2001
2002
3649
3650


590
2003
2004
3651
3652


591
2005
2006
3653
3654


592
2007
2008
3655
3656


593
2009
2010
3657
3658


594
2011
2012
3659
3660


595
2013
2014
3661
3662


596
2015
2016
3663
3664


597
2017
2018
3665
3666


598
2019
2020
3667
3668


599
2021
2022
3669
3670


600
2023
2024
3671
3672


601
2025
2026
3673
3674


602
2027
2028
3675
3676


603
2029
2030
3677
3678


604
2031
2032
3679
3680


605
2033
2034
3681
3682


606
2035
2036
3683
3684


607
2037
2038
3685
3686


608
2039
2040
3687
3688


609
2041
2042
3689
3690


610
2043
2044
3691
3692


611
2045
2046
3693
3694


612
2047
2048
3695
3696


613
2049
2050
3697
3698


614
2051
2052
3699
3700


615
2053
2054
3701
3702


616
2055
2056
3703
3704


617
2057
2058
3705
3706


618
2059
2060
3707
3708


619
2061
2062
3709
3710


620
2063
2064
3711
3712


621
2065
2066
3713
3714


622
2067
2068
3715
3716


623
2069
2070
3717
3718


624
2071
2072
3719
3720


625
2073
2074
3721
3722


626
2075
2076
3723
3724


627
2077
2078
3725
3726


628
2079
2080
3727
3728


629
2081
2082
3729
3730


630
2083
2084
3731
3732


631
2085
2086
3733
3734


632
2087
2088
3735
3736


633
2089
2090
3737
3738


634
2091
2092
3739
3740


635
2093
2094
3741
3742


636
2095
2096
3743
3744


637
2097
2098
3745
3746


638
2099
2100
3747
3748


639
2101
2102
3749
3750


640
2103
2104
3751
3752


641
2105
2106
3753
3754


642
2107
2108
3755
3756


643
2109
2110
3757
3758


644
2111
2112
3759
3760


645
2113
2114
3761
3762


646
2115
2116
3763
3764


647
2117
2118
3765
3766


648
2119
2120
3767
3768


649
2121
2122
3769
3770


650
2123
2124
3771
3772


651
2125
2126
3773
3774


652
2127
2128
3775
3776


653
2129
2130
3777
3778


654
2131
2132
3779
3780


655
2133
2134
3781
3782


656
2135
2136
3783
3784


657
2137
2138
3785
3786


658
2139
2140
3787
3788


659
2141
2142
3789
3790


660
2143
2144
3791
3792


661
2145
2146
3793
3794


662
2147
2148
3795
3796


663
2149
2150
3797
3798


664
2151
2152
3799
3800


665
2153
2154
3801
3802


666
2155
2156
3803
3804


667
2157
2158
3805
3806


668
2159
2160
3807
3808


669
2161
2162
3809
3810


670
2163
2164
3811
3812


671
2165
2166
3813
3814


672
2167
2168
3815
3816


673
2169
2170
3817
3818


674
2171
2172
3819
3820


675
2173
2174
3821
3822


676
2175
2176
3823
3824


677
2177
2178
3825
3826


678
2179
2180
3827
3828


679
2181
2182
3829
3830


680
2183
2184
3831
3832


681
2185
2186
3833
3834


682
2187
2188
3835
3836


683
2189
2190
3837
3838


684
2191
2192
3839
3840


685
2193
2194
3841
3842


686
2195
2196
3843
3844


687
2197
2198
3845
3846


688
2199
2200
3847
3848


689
2201
2202
3849
3850


690
2203
2204
3851
3852


691
2205
2206
3853
3854


692
2207
2208
3855
3856


693
2209
2210
3857
3858


694
2211
2212
3859
3860


695
2213
2214
3861
3862


696
2215
2216
3863
3864


697
2217
2218
3865
3866


698
2219
2220
3867
3868


699
2221
2222
3869
3870


700
2223
2224
3871
3872


701
2225
2226
3873
3874


702
2227
2228
3875
3876


703
2229
2230
3877
3878


704
2231
2232
3879
3880


705
2233
2234
3881
3882


706
2235
2236
3883
3884


707
2237
2238
3885
3886


708
2239
2240
3887
3888


709
2241
2242
3889
3890


710
2243
2244
3891
3892


711
2245
2246
3893
3894


712
2247
2248
3895
3896


713
2249
2250
3897
3898


714
2251
2252
3899
3900


715
2253
2254
3901
3902


716
2255
2256
3903
3904


717
2257
2258
3905
3906


718
2259
2260
3907
3908


719
2261
2262
3909
3910


720
2263
2264
3911
3912


721
2265
2266
3913
3914


722
2267
2268
3915
3916


723
2269
2270
3917
3918


724
2271
2272
3919
3920


725
2273
2274
3921
3922


726
2275
2276
3923
3924


727
2277
2278
3925
3926


728
2279
2280
3927
3928


729
2281
2282
3929
3930


730
2283
2284
3931
3932


731
2285
2286
3933
3934


732
2287
2288
3935
3936


733
2289
2290
3937
3938


734
2291
2292
3939
3940


735
2293
2294
3941
3942


736
2295
2296
3943
3944


737
2297
2298
3945
3946


738
2299
2300
3947
3948


739
2301
2302
3949
3950


740
2303
2304
3951
3952


741
2305
2306
3953
3954


742
2307
2308
3955
3956


743
2309
2310
3957
3958


744
2311
2312
3959
3960


745
2313
2314
3961
3962


746
2315
2316
3963
3964


747
2317
2318
3965
3966


748
2319
2320
3967
3968


749
2321
2322
3969
3970


750
2323
2324
3971
3972


751
2325
2326
3973
3974


752
2327
2328
3975
3976


753
2329
2330
3977
3978


754
2331
2332
3979
3980


755
2333
2334
3981
3982


756
2335
2336
3983
3984


757
2337
2338
3985
3986


758
2339
2340
3987
3988


759
2341
2342
3989
3990


760
2343
2344
3991
3992


761
2345
2346
3993
3994


762
2347
2348
3995
3996


763
2349
2350
3997
3998


764
2351
2352
3999
4000


765
2353
2354
4001
4002


766
2355
2356
4003
4004


767
2357
2358
4005
4006


768
2359
2360
4007
4008


769
2361
2362
4009
4010


770
2363
2364
4011
4012


771
2365
2366
4013
4014


772
2367
2368
4015
4016


773
2369
2370
4017
4018


774
2371
2372
4019
4020


775
2373
2374
4021
4022


776
2375
2376
4023
4024


777
2377
2378
4025
4026


778
2379
2380
4027
4028


779
2381
2382
4029
4030


780
2383
2384
4031
4032


781
2385
2386
4033
4034


782
2387
2388
4035
4036


783
2389
2390
4037
4038


784
2391
2392
4039
4040


785
2393
2394
4041
4042


786
2395
2396
4043
4044


787
2397
2398
4045
4046


788
2399
2400
4047
4048


789
2401
2402
4049
4050


790
2403
2404
4051
4052


791
2405
2406
4053
4054


792
2407
2408
4055
4056


793
2409
2410
4057
4058


794
2411
2412
4059
4060


795
2413
2414
4061
4062


796
2415
2416
4063
4064


797
2417
2418
4065
4066


798
2419
2420
4067
4068


799
2421
2422
4069
4070


800
2423
2424
4071
4072


801
2425
2426
4073
4074


802
2427
2428
4075
4076


803
2429
2430
4077
4078


804
2431
2432
4079
4080


805
2433
2434
4081
4082


806
2435
2436
4083
4084


807
2437
2438
4085
4086


808
2439
2440
4087
4088


809
2441
2442
4089
4090


810
2443
2444
4091
4092


811
2445
2446
4093
4094


812
2447
2448
4095
4096


813
2449
2450
4097
4098


814
2451
2452
4099
4100


815
2453
2454
4101
4102


816
2455
2456
4103
4104


817
2457
2458
4105
4106


818
2459
2460
4107
4108


819
2461
2462
4109
4110


820
2463
2464
4111
4112


821
2465
2466
4113
4114


822
2467
2468
4115
4116


823
2469
2470
4117
4118


824
2471
2472
4119
4120
















TABLE 3







Overview of SEQ ID NO of amplificates derived from the genes or


genomic regions and the SEQ ID NOs of corresponding bisulfite treated


nucleic acids (DNA upmethylated sense strand, DNA upmethylated


antisense strand, DNA downmethylated sense strand, DNA


downmethylated antisense strand).














SEQ ID NO:
SEQ ID NO:


SEQ ID
SEQ ID NO:
SEQ ID NO:
Down-
Down-


NO:
Upmethylated
Upmethylated
methylated
methylated


Genomic
Sense
Antisense
Sense
Antisense














1
825
826
2473
2474


2
827
828
2475
2476


3
829
830
2477
2478


4
831
832
2479
2480


5
833
834
2481
2482


6
835
836
2483
2484


7
837
838
2485
2486


8
839
840
2487
2488


9
841
842
2489
2490


10
843
844
2491
2492


11
845
846
2493
2494


12
847
848
2495
2496


13
849
850
2497
2498


14
851
852
2499
2500


15
853
854
2501
2502


16
855
856
2503
2504


17
857
858
2505
2506


18
859
860
2507
2508


19
861
862
2509
2510


20
863
864
2511
2512


21
865
866
2513
2514


22
867
868
2515
2516


23
869
870
2517
2518


24
871
872
2519
2520


25
873
874
2521
2522


26
875
876
2523
2524


27
877
878
2525
2526


28
879
880
2527
2528


29
881
882
2529
2530


30
883
884
2531
2532


31
885
886
2533
2534


32
887
888
2535
2536


33
889
890
2537
2538


34
891
892
2539
2540


35
893
894
2541
2542


36
895
896
2543
2544


37
897
898
2545
2546


38
899
900
2547
2548


39
901
902
2549
2550


40
903
904
2551
2552


41
905
906
2553
2554


42
907
908
2555
2556


43
909
910
2557
2558


44
911
912
2559
2560


45
913
914
2561
2562


46
915
916
2563
2564


47
917
918
2565
2566


48
919
920
2567
2568


49
921
922
2569
2570


50
923
924
2571
2572


51
925
926
2573
2574


52
927
928
2575
2576


53
929
930
2577
2578


54
931
932
2579
2580


55
933
934
2581
2582


56
935
936
2583
2584


57
937
938
2585
2586


58
939
940
2587
2588


59
941
942
2589
2590


60
943
944
2591
2592


61
945
946
2593
2594


62
947
948
2595
2596


63
949
950
2597
2598


64
951
952
2599
2600


65
953
954
2601
2602


66
955
956
2603
2604


67
957
958
2605
2606


68
959
960
2607
2608


69
961
962
2609
2610


70
963
964
2611
2612


71
965
966
2613
2614


72
967
968
2615
2616


73
969
970
2617
2618


74
971
972
2619
2620


75
973
974
2621
2622


76
975
976
2623
2624


77
977
978
2625
2626


78
979
980
2627
2628


79
981
982
2629
2630


80
983
984
2631
2632


81
985
986
2633
2634


82
987
988
2635
2636


83
989
990
2637
2638


84
991
992
2639
2640


85
993
994
2641
2642


86
995
996
2643
2644


87
997
998
2645
2646


88
999
1000
2647
2648


89
1001
1002
2649
2650


90
1003
1004
2651
2652


91
1005
1006
2653
2654


92
1007
1008
2655
2656


93
1009
1010
2657
2658


94
1011
1012
2659
2660


95
1013
1014
2661
2662


96
1015
1016
2663
2664


97
1017
1018
2665
2666


98
1019
1020
2667
2668


99
1021
1022
2669
2670


100
1023
1024
2671
2672


101
1025
1026
2673
2674


102
1027
1028
2675
2676


103
1029
1030
2677
2678


104
1031
1032
2679
2680


105
1033
1034
2681
2682


106
1035
1036
2683
2684


107
1037
1038
2685
2686


108
1039
1040
2687
2688


109
1041
1042
2689
2690


110
1043
1044
2691
2692


111
1045
1046
2693
2694


112
1047
1048
2695
2696


113
1049
1050
2697
2698


114
1051
1052
2699
2700


115
1053
1054
2701
2702


116
1055
1056
2703
2704


117
1057
1058
2705
2706


118
1059
1060
2707
2708


119
1061
1062
2709
2710


120
1063
1064
2711
2712


121
1065
1066
2713
2714


122
1067
1068
2715
2716


123
1069
1070
2717
2718


124
1071
1072
2719
2720


125
1073
1074
2721
2722


126
1075
1076
2723
2724


127
1077
1078
2725
2726


128
1079
1080
2727
2728


129
1081
1082
2729
2730


130
1083
1084
2731
2732


131
1085
1086
2733
2734


132
1087
1088
2735
2736


133
1089
1090
2737
2738


134
1091
1092
2739
2740


135
1093
1094
2741
2742


136
1095
1096
2743
2744


137
1097
1098
2745
2746


138
1099
1100
2747
2748


139
1101
1102
2749
2750


140
1103
1104
2751
2752


141
1105
1106
2753
2754


142
1107
1108
2755
2756


143
1109
1110
2757
2758


144
1111
1112
2759
2760


145
1113
1114
2761
2762


146
1115
1116
2763
2764


147
1117
1118
2765
2766


148
1119
1120
2767
2768


149
1121
1122
2769
2770


150
1123
1124
2771
2772


151
1125
1126
2773
2774


152
1127
1128
2775
2776


153
1129
1130
2777
2778


154
1131
1132
2779
2780


155
1133
1134
2781
2782


156
1135
1136
2783
2784


157
1137
1138
2785
2786


158
1139
1140
2787
2788


159
1141
1142
2789
2790


160
1143
1144
2791
2792


161
1145
1146
2793
2794


162
1147
1148
2795
2796


163
1149
1150
2797
2798


164
1151
1152
2799
2800


165
1153
1154
2801
2802


166
1155
1156
2803
2804


167
1157
1158
2805
2806


168
1159
1160
2807
2808


169
1161
1162
2809
2810


170
1163
1164
2811
2812


171
1165
1166
2813
2814


172
1167
1168
2815
2816


173
1169
1170
2817
2818


174
1171
1172
2819
2820


175
1173
1174
2821
2822


176
1175
1176
2823
2824


177
1177
1178
2825
2826


178
1179
1180
2827
2828


179
1181
1182
2829
2830


180
1183
1184
2831
2832


181
1185
1186
2833
2834


182
1187
1188
2835
2836


183
1189
1190
2837
2838


184
1191
1192
2839
2840


185
1193
1194
2841
2842


186
1195
1196
2843
2844


187
1197
1198
2845
2846


188
1199
1200
2847
2848


189
1201
1202
2849
2850


190
1203
1204
2851
2852


191
1205
1206
2853
2854


192
1207
1208
2855
2856


193
1209
1210
2857
2858


194
1211
1212
2859
2860


195
1213
1214
2861
2862


196
1215
1216
2863
2864


197
1217
1218
2865
2866


198
1219
1220
2867
2868


199
1221
1222
2869
2870


200
1223
1224
2871
2872


201
1225
1226
2873
2874


202
1227
1228
2875
2876


203
1229
1230
2877
2878


204
1231
1232
2879
2880


205
1233
1234
2881
2882


206
1235
1236
2883
2884


207
1237
1238
2885
2886


208
1239
1240
2887
2888


209
1241
1242
2889
2890


210
1243
1244
2891
2892


211
1245
1246
2893
2894


212
1247
1248
2895
2896


213
1249
1250
2897
2898


214
1251
1252
2899
2900


215
1253
1254
2901
2902


216
1255
1256
2903
2904


217
1257
1258
2905
2906


218
1259
1260
2907
2908


219
1261
1262
2909
2910


220
1263
1264
2911
2912


221
1265
1266
2913
2914


222
1267
1268
2915
2916


223
1269
1270
2917
2918


224
1271
1272
2919
2920


225
1273
1274
2921
2922


226
1275
1276
2923
2924


227
1277
1278
2925
2926


228
1279
1280
2927
2928


229
1281
1282
2929
2930


230
1283
1284
2931
2932


231
1285
1286
2933
2934


232
1287
1288
2935
2936


233
1289
1290
2937
2938


234
1291
1292
2939
2940


235
1293
1294
2941
2942


236
1295
1296
2943
2944


237
1297
1298
2945
2946


238
1299
1300
2947
2948


239
1301
1302
2949
2950


240
1303
1304
2951
2952


241
1305
1306
2953
2954


242
1307
1308
2955
2956


243
1309
1310
2957
2958


244
1311
1312
2959
2960


245
1313
1314
2961
2962


246
1315
1316
2963
2964


247
1317
1318
2965
2966


248
1319
1320
2967
2968


249
1321
1322
2969
2970


250
1323
1324
2971
2972


251
1325
1326
2973
2974


252
1327
1328
2975
2976


253
1329
1330
2977
2978


254
1331
1332
2979
2980


255
1333
1334
2981
2982


256
1335
1336
2983
2984


257
1337
1338
2985
2986


258
1339
1340
2987
2988


259
1341
1342
2989
2990


260
1343
1344
2991
2992


261
1345
1346
2993
2994


262
1347
1348
2995
2996


263
1349
1350
2997
2998


264
1351
1352
2999
3000


265
1353
1354
3001
3002


266
1355
1356
3003
3004


267
1357
1358
3005
3006


268
1359
1360
3007
3008


269
1361
1362
3009
3010


270
1363
1364
3011
3012


271
1365
1366
3013
3014


272
1367
1368
3015
3016


273
1369
1370
3017
3018


274
1371
1372
3019
3020


275
1373
1374
3021
3022


276
1375
1376
3023
3024


277
1377
1378
3025
3026


278
1379
1380
3027
3028


279
1381
1382
3029
3030


280
1383
1384
3031
3032


281
1385
1386
3033
3034


282
1387
1388
3035
3036


283
1389
1390
3037
3038


284
1391
1392
3039
3040


285
1393
1394
3041
3042


286
1395
1396
3043
3044


287
1397
1398
3045
3046


288
1399
1400
3047
3048


289
1401
1402
3049
3050


290
1403
1404
3051
3052


291
1405
1406
3053
3054


292
1407
1408
3055
3056


293
1409
1410
3057
3058


294
1411
1412
3059
3060


295
1413
1414
3061
3062


296
1415
1416
3063
3064


297
1417
1418
3065
3066


298
1419
1420
3067
3068


299
1421
1422
3069
3070


300
1423
1424
3071
3072


301
1425
1426
3073
3074


302
1427
1428
3075
3076


303
1429
1430
3077
3078


304
1431
1432
3079
3080


305
1433
1434
3081
3082


306
1435
1436
3083
3084


307
1437
1438
3085
3086


308
1439
1440
3087
3088


309
1441
1442
3089
3090


310
1443
1444
3091
3092


311
1445
1446
3093
3094


312
1447
1448
3095
3096


313
1449
1450
3097
3098


314
1451
1452
3099
3100


315
1453
1454
3101
3102


316
1455
1456
3103
3104


317
1457
1458
3105
3106


318
1459
1460
3107
3108


319
1461
1462
3109
3110


320
1463
1464
3111
3112


321
1465
1466
3113
3114


322
1467
1468
3115
3116


323
1469
1470
3117
3118


324
1471
1472
3119
3120


325
1473
1474
3121
3122


326
1475
1476
3123
3124


327
1477
1478
3125
3126


328
1479
1480
3127
3128


329
1481
1482
3129
3130


330
1483
1484
3131
3132


331
1485
1486
3133
3134


332
1487
1488
3135
3136


333
1489
1490
3137
3138


334
1491
1492
3139
3140


335
1493
1494
3141
3142


336
1495
1496
3143
3144


337
1497
1498
3145
3146


338
1499
1500
3147
3148


339
1501
1502
3149
3150


340
1503
1504
3151
3152


341
1505
1506
3153
3154


342
1507
1508
3155
3156


343
1509
1510
3157
3158


344
1511
1512
3159
3160


345
1513
1514
3161
3162


346
1515
1516
3163
3164


347
1517
1518
3165
3166


348
1519
1520
3167
3168


349
1521
1522
3169
3170


350
1523
1524
3171
3172


351
1525
1526
3173
3174


352
1527
1528
3175
3176


353
1529
1530
3177
3178


354
1531
1532
3179
3180


355
1533
1534
3181
3182


356
1535
1536
3183
3184


357
1537
1538
3185
3186


358
1539
1540
3187
3188


359
1541
1542
3189
3190


360
1543
1544
3191
3192


361
1545
1546
3193
3194


362
1547
1548
3195
3196


363
1549
1550
3197
3198


364
1551
1552
3199
3200


365
1553
1554
3201
3202


366
1555
1556
3203
3204


367
1557
1558
3205
3206


368
1559
1560
3207
3208


369
1561
1562
3209
3210


370
1563
1564
3211
3212


371
1565
1566
3213
3214


372
1567
1568
3215
3216


373
1569
1570
3217
3218


374
1571
1572
3219
3220


375
1573
1574
3221
3222


376
1575
1576
3223
3224


377
1577
1578
3225
3226


378
1579
1580
3227
3228


379
1581
1582
3229
3230


380
1583
1584
3231
3232


381
1585
1586
3233
3234


382
1587
1588
3235
3236


383
1589
1590
3237
3238


384
1591
1592
3239
3240


385
1593
1594
3241
3242


386
1595
1596
3243
3244


387
1597
1598
3245
3246


388
1599
1600
3247
3248


389
1601
1602
3249
3250


390
1603
1604
3251
3252


391
1605
1606
3253
3254


392
1607
1608
3255
3256


393
1609
1610
3257
3258


394
1611
1612
3259
3260


395
1613
1614
3261
3262


396
1615
1616
3263
3264


397
1617
1618
3265
3266


398
1619
1620
3267
3268


399
1621
1622
3269
3270


400
1623
1624
3271
3272


401
1625
1626
3273
3274


402
1627
1628
3275
3276


403
1629
1630
3277
3278


404
1631
1632
3279
3280


405
1633
1634
3281
3282


406
1635
1636
3283
3284


407
1637
1638
3285
3286


408
1639
1640
3287
3288


409
1641
1642
3289
3290


410
1643
1644
3291
3292


411
1645
1646
3293
3294


412
1647
1648
3295
3296
















TABLE 4







Overview of SEQ ID NO of the genes or genomic regions and of


corresponding amplificates amplified by use of the corresponding


Forward and Reverse Primer named by their SEQ ID NO.












SEQ
Forward Primer
Reverse Primer
SEQ



ID NO:
SEQ ID
SEQ ID
ID NO:



Genomic
NO:
NO:
Genomic
















413
4121
4122
1



414
4123
4124
2



415
4125
4126
3



416
4127
4128
4



417
4129
4130
5



418
4131
4132
6



419
4133
4134
7



420
4135
4136
8



421
4137
4138
9



422
4139
4140
10



423
4141
4142
11



424
4143
4144
12



425
4145
4146
13



426
4147
4148
14



427
4149
4150
15



428
4151
4152
16



429
4153
4154
17



430
4155
4156
18



431
4157
4158
19



432
4159
4160
20



433
4161
4162
21



434
4163
4164
22



435
4165
4166
23



436
4167
4168
24



437
4169
4170
25



438
4171
4172
26



439
4173
4174
27



440
4175
4176
28



441
4177
4178
29



442
4179
4180
30



443
4181
4182
31



444
4183
4184
32



445
4185
4186
33



446
4187
4188
34



447
4189
4190
35



448
4191
4192
36



449
4193
4194
37



450
4195
4196
38



451
4197
4198
39



452
4199
4200
40



453
4201
4202
41



454
4203
4204
42



455
4205
4206
43



456
4207
4208
44



457
4209
4210
45



458
4211
4212
46



459
4213
4214
47



460
4215
4216
48



461
4217
4218
49



462
4219
4220
50



463
4221
4222
51



464
4223
4224
52



465
4225
4226
53



466
4227
4228
54



467
4229
4230
55



468
4231
4232
56



469
4233
4234
57



470
4235
4236
58



471
4237
4238
59



472
4239
4240
60



473
4241
4242
61



474
4243
4244
62



475
4245
4246
63



476
4247
4248
64



477
4249
4250
65



478
4251
4252
66



479
4253
4254
67



480
4255
4256
68



481
4257
4258
69



482
4259
4260
70



483
4261
4262
71



484
4263
4264
72



485
4265
4266
73



486
4267
4268
74



487
4269
4270
75



488
4271
4272
76



489
4273
4274
77



490
4275
4276
78



491
4277
4278
79



492
4279
4280
80



493
4281
4282
81



494
4283
4284
82



495
4285
4286
83



496
4287
4288
84



497
4289
4290
85



498
4291
4292
86



499
4293
4294
87



500
4295
4296
88



501
4297
4298
89



502
4299
4300
90



503
4301
4302
91



504
4303
4304
92



505
4305
4306
93



506
4307
4308
94



507
4309
4310
95



508
4311
4312
96



509
4313
4314
97



510
4315
4316
98



511
4317
4318
99



512
4319
4320
100



513
4321
4322
101



514
4323
4324
102



515
4325
4326
103



516
4327
4328
104



517
4329
4330
105



518
4331
4332
106



519
4333
4334
107



520
4335
4336
108



521
4337
4338
109



522
4339
4340
110



523
4341
4342
111



524
4343
4344
112



525
4345
4346
113



526
4347
4348
114



527
4349
4350
115



528
4351
4352
116



529
4353
4354
117



530
4355
4356
118



531
4357
4358
119



532
4359
4360
120



533
4361
4362
121



534
4363
4364
122



535
4365
4366
123



536
4367
4368
124



537
4369
4370
125



538
4371
4372
126



539
4373
4374
127



540
4375
4376
128



541
4377
4378
129



542
4379
4380
130



543
4381
4382
131



544
4383
4384
132



545
4385
4386
133



546
4387
4388
134



547
4389
4390
135



548
4391
4392
136



549
4393
4394
137



550
4395
4396
138



551
4397
4398
139



552
4399
4400
140



553
4401
4402
141



554
4403
4404
142



555
4405
4406
143



556
4407
4408
144



557
4409
4410
145



558
4411
4412
146



559
4413
4414
147



560
4415
4416
148



561
4417
4418
149



562
4419
4420
150



563
4421
4422
151



564
4423
4424
152



565
4425
4426
153



566
4427
4428
154



567
4429
4430
155



568
4431
4432
156



569
4433
4434
157



570
4435
4436
158



571
4437
4438
159



572
4439
4440
160



573
4441
4442
161



574
4443
4444
162



575
4445
4446
163



576
4447
4448
164



577
4449
4450
165



578
4451
4452
166



579
4453
4454
167



580
4455
4456
168



581
4457
4458
169



582
4459
4460
170



583
4461
4462
171



584
4463
4464
172



585
4465
4466
173



586
4467
4468
174



587
4469
4470
175



588
4471
4472
176



589
4473
4474
177



590
4475
4476
178



591
4477
4478
179



592
4479
4480
180



593
4481
4482
181



594
4483
4484
182



595
4485
4486
183



596
4487
4488
184



597
4489
4490
185



598
4491
4492
186



599
4493
4494
187



600
4495
4496
188



601
4497
4498
189



602
4499
4500
190



603
4501
4502
191



604
4503
4504
192



605
4505
4506
193



606
4507
4508
194



607
4509
4510
195



608
4511
4512
196



609
4513
4514
197



610
4515
4516
198



611
4517
4518
199



612
4519
4520
200



613
4521
4522
201



614
4523
4524
202



615
4525
4526
203



616
4527
4528
204



617
4529
4530
205



618
4531
4532
206



619
4533
4534
207



620
4535
4536
208



621
4537
4538
209



622
4539
4540
210



623
4541
4542
211



624
4543
4544
212



625
4545
4546
213



626
4547
4548
214



627
4549
4550
215



628
4551
4552
216



629
4553
4554
217



630
4555
4556
218



631
4557
4558
219



632
4559
4560
220



633
4561
4562
221



634
4563
4564
222



635
4565
4566
223



636
4567
4568
224



637
4569
4570
225



638
4571
4572
226



639
4573
4574
227



640
4575
4576
228



641
4577
4578
229



642
4579
4580
230



643
4581
4582
231



644
4583
4584
232



645
4585
4586
233



646
4587
4588
234



647
4589
4590
235



648
4591
4592
236



649
4593
4594
237



650
4595
4596
238



651
4597
4598
239



652
4599
4600
240



653
4601
4602
241



654
4603
4604
242



655
4605
4606
243



656
4607
4608
244



657
4609
4610
245



658
4611
4612
246



659
4613
4614
247



660
4615
4616
248



661
4617
4618
249



662
4619
4620
250



663
4621
4622
251



664
4623
4624
252



665
4625
4626
253



666
4627
4628
254



667
4629
4630
255



668
4631
4632
256



669
4633
4634
257



670
4635
4636
258



671
4637
4638
259



672
4639
4640
260



673
4641
4642
261



674
4643
4644
262



675
4645
4646
263



676
4647
4648
264



677
4649
4650
265



678
4651
4652
266



679
4653
4654
267



680
4655
4656
268



681
4657
4658
269



682
4659
4660
270



683
4661
4662
271



684
4663
4664
272



685
4665
4666
273



686
4667
4668
274



687
4669
4670
275



688
4671
4672
276



689
4673
4674
277



690
4675
4676
278



691
4677
4678
279



692
4679
4680
280



693
4681
4682
281



694
4683
4684
282



695
4685
4686
283



696
4687
4688
284



697
4689
4690
285



698
4691
4692
286



699
4693
4694
287



700
4695
4696
288



701
4697
4698
289



702
4699
4700
290



703
4701
4702
291



704
4703
4704
292



705
4705
4706
293



706
4707
4708
294



707
4709
4710
295



708
4711
4712
296



709
4713
4714
297



710
4715
4716
298



711
4717
4718
299



712
4719
4720
300



713
4721
4722
301



714
4723
4724
302



715
4725
4726
303



716
4727
4728
304



717
4729
4730
305



718
4731
4732
306



719
4733
4734
307



720
4735
4736
308



721
4737
4738
309



722
4739
4740
310



723
4741
4742
311



724
4743
4744
312



725
4745
4746
313



726
4747
4748
314



727
4749
4750
315



728
4751
4752
316



729
4753
4754
317



730
4755
4756
318



731
4757
4758
319



732
4759
4760
320



733
4761
4762
321



734
4763
4764
322



735
4765
4766
323



736
4767
4768
324



737
4769
4770
325



738
4771
4772
326



739
4773
4774
327



740
4775
4776
328



741
4777
4778
329



742
4779
4780
330



743
4781
4782
331



744
4783
4784
332



745
4785
4786
333



746
4787
4788
334



747
4789
4790
335



748
4791
4792
336



749
4793
4794
337



750
4795
4796
338



751
4797
4798
339



752
4799
4800
340



753
4801
4802
341



754
4803
4804
342



755
4805
4806
343



756
4807
4808
344



757
4809
4810
345



758
4811
4812
346



759
4813
4814
347



760
4815
4816
348



761
4817
4818
349



762
4819
4820
350



763
4821
4822
351



764
4823
4824
352



765
4825
4826
353



766
4827
4828
354



767
4829
4830
355



768
4831
4832
356



769
4833
4834
357



770
4835
4836
358



771
4837
4838
359



772
4839
4840
360



773
4841
4842
361



774
4843
4844
362



775
4845
4846
363



776
4847
4848
364



777
4849
4850
365



778
4851
4852
366



779
4853
4854
367



780
4855
4856
368



781
4857
4858
369



782
4859
4860
370



783
4861
4862
371



784
4863
4864
372



785
4865
4866
373



786
4867
4868
374



787
4869
4870
375



788
4871
4872
376



789
4873
4874
377



790
4875
4876
378



791
4877
4878
379



792
4879
4880
380



793
4881
4882
381



794
4883
4884
382



795
4885
4886
383



796
4887
4888
384



797
4889
4890
385



798
4891
4892
386



799
4893
4894
387



800
4895
4896
388



801
4897
4898
389



802
4899
4900
390



803
4901
4902
391



804
4903
4904
392



805
4905
4906
393



806
4907
4908
394



807
4909
4910
395



808
4911
4912
396



809
4913
4914
397



810
4915
4916
398



811
4917
4918
399



812
4919
4920
400



813
4921
4922
401



814
4923
4924
402



815
4925
4926
403



816
4927
4928
404



817
4929
4930
405



818
4931
4932
406



819
4933
4934
407



820
4935
4936
408



821
4937
4938
409



822
4939
4940
410



823
4941
4942
411



824
4943
4944
412

















TABLE 5







Overview of genes or genomic regions and the corresponding SEQ ID NOs, HUGO IDs,


Aliases, Reference Sequence IDs, Ensemble Gene IDs and Entrez IDs.













SEQ








ID


NO:


Ge-


nom-




Ensembl Gene
Entrez Gene


ic
Gene
HUGO ID
Aliases
RefSeq ID
ID
ID
















413
FLOT1, flotillin 1,
FLOT1

NM_005803
ENSG00000137312
10211



ENSG00000137312


414
C6orf25, chromosome
C6orf25
G6b;
NM_025260
ENSG00000096148
80739



6 open reading frame

NG31



25,



ENSG00000096148


415
VARS, valyl-tRNA
VARS
G7A;
NM_006295
ENSG00000096171
7407



synthetase,

VARS2



ENSG00000096171


416
major
HLA-
DPB1;
NM_002121
OTTHUMG00000031076
3115



histocompatibility
DPB1
HLA-



complex, class II, DP

DP1B;



beta 1,

MHC



OTTHUMG00000031076,

DPB1



HLA-DPB1


417
HLA-DRB5, major
HLA-

NG_002432
OTTHUMG00000031027
3127



histocompatibility
DRB5



complex, class II, DR



beta 5,



OTTHUMG00000031027


418
COL11A2, collagen,
COL11A2
HKE5;
NM_080679
OTTHUMG00000031036
1302



type XI, alpha 2,

PARP;



OTTHUMG00000031036

STL3;





DFNA13;





DFNB53


419
PRAME, Melanoma
PRAME
MAPE;
NG_000002
ENSG00000185686
23532



antigen preferentially

OIP4



expressed in tumors



(Preferentially



expressed antigen of



melanoma) (OPA-



interacting protein 4)



(OIP4), ENSG00000185686


420
ZNRF3 protein
ZNRF3
KIAA1133,
XM_290972,
ENSG00000183579
84133



(Fragment),

BK747E2.3,
XP_290972



ENSG00000183579,

FLJ22057,



ZNRF3 zinc and ring

RNF203



finger 3 (ZNRF3)


421
AP000357.2 (Vega
AP000357.2
Em: AP000357.C22.2
No
OTTHUMG00000030571
No Available



gene ID), Pseudogene
(Vega

Available




gene ID)


422
AP000357.3 (Vega
AP000357.3
Em: AP000357.C22.3
No
OTTHUMG00000030574
No Available



gene ID), Pseudogene
(Vega

Available




gene ID)


423
solute carrier family 7
SLC7A4
CAT4;
NM_004173
OTTHUMG00000030129
6545



(cationic amino acid

CAT-4;



transporter, y+

HCAT3;



system), member 4,

MGC129976;



OTTHUMG00000030129,

MGC129977



SLC7A4 (


424
Myosin-18B (Myosin
MYO18B
BK125H2.1
NM_014550
ENSG00000133454
84700



XVIIIb),



ENSG00000133454,



MYO18B


425
Q6ICL0_HUMAN
Q6ICL0_HUMAN
Em: AC006547.7,

ENSG00000184004
150197



(Predicted
(Predicted
FLJ32575



UniProt/TrEMBL ID),
UniProt/TrEMBL



hypothetical protein
ID)



FLJ3257;



ENSG00000184004


426
FBLN1; fibulin 1;
FBLN1
FBLN
NM_001996
ENSG00000077942
2192



ENSG00000077942


427
CYP2D6; cytochrome
CYP2D6
CPD6;
NG_003180
ENSG00000100197
1565



P450, family 2,

CYP2D;



subfamily D,

CYP2D@;



polypeptide 6;

CYP2DL1;



ENSG00000100197

450C2D;





P450-DB1;





MGC120389;





MGC120390


428
AC008132.9 (Vega
AC008132.9
AC008103.3;
No
OTTHUMG00000030688
No Available



gene ID); Pseudogene;
(Vega
Em: AC008132.C22.2
Available



OTTHUMG00000030688
gene ID)


429
glycoprotein Ib
GP1BB

NM_000407
OTTHUMT00000075045
2812



(platelet), beta



polypeptide,



OTTHUMT00000075045,



GP1BB-001


430
no gene associated


431
AC006548.8 (Vega
AC006548.8
Em: AC006548.C22.8
No
OTTHUMG00000030274
No Available



gene ID)
(Vega

Available




gene ID)


432
OTTHUMG00000030650,
AC005399.2
Em: AC005399.C22.2
No
OTTHUMG00000030650
No Available



AC005399.2,
(Vega

Available



putativer processed
gene ID)



transcribed


433
topoisomerase (DNA)
TOP3B

NM_003935
OTTHUMG00000030764
8940



III beta,



OTTHUMG00000030764,



TOP3B (


434
no gene associated


435
KB-1269D1.3 (Vega
KB-
Em: AP000344.C22.3
No
OTTHUMG00000030694
No Available



gene ID); Pseudogene;
1269D1.3

Available



OTTHUMG00000030694
(Vega gene




ID)


436
GPR24; G protein-
GPR24
SLC1;
NM_005297
ENSG00000128285
2847



coupled receptor 24;

MCHR1;



ENSG00000128285

MGC32129


437
GAL3ST1; galactose-
GAL3ST1
CST
NM_004861
ENSG00000128242
9514



3-O-sulfotransferase 1;



ENSG00000128242


438
Cat eye syndrome
CECR5

NM_017829
ENSG00000069998
27440



critical region protein



5 precursor,



ENSG00000069998,



CECR5 and (head to



head)



OTTHUMG00000030478,



AC006946.7


439
HORMAD2; HORMA
HORMAD2
MGC26710
NM_152510
ENSG00000176635
150280



domain containing 2;



ENSG00000176635


440
OTTHUMG00000030922,
RP3-
Em: AC002378.C22.2
No
OTTHUMG00000030922
No Available



RP3-438O4.2
438O4.2

Available




(Vega_gene




ID)


441
NP_997357.1 (RefSeq
NP_997357.1
FLJ42953
NM_207474
ENSG00000169668
400892



peptide ID);
(RefSeq



ENSG00000169668
peptide ID)


442
OTTHUMG00000030574,
AP000357.3
Em: AP000357.C22.3

OTTHUMG00000030574



AP000357.3,



novel pseudogene


443
LA16c-4G1.2 (Vega
LA16c-
Em: AP000522.C22.2
No
OTTHUMG00000030832
No Available



gene ID); Pseudogene;
4G1.2

Available



OTTHUMG00000030832
(Vega gene




ID)


444
KB-226F1.11 (Vega
KB-
Em:
No
OTTHUMG00000030123
No Available



gene ID), embryonic
226F1.11
AP000351.C22.11
Available



marker,
(Vega gene



OTTHUMG00000030123
ID)


445
OTTHUMG00000030780,
CTA-
bK373H7.C22.4
No
OTTHUMG00000030780
No Available



CTA-373H7.4,
373H7.4

Available



novel pseudogene
(Vega gene




ID)


446
RP1-47A17.8 (Vega
RP1-
dJ47A17.C22.8
No
OTTHUMG00000030878
No Available



gene ID);
47A17.8

Available



OTTHUMG00000030878
(Vega gene




ID);


447
RP4-539M6.7 (Vega
RP4-
Em: AC004832.C22.7
No
OTTHUMG00000030918
No Available



gene ID); Pseudogene;
539M6.7

Available



OTTHUMG00000030918
(Vega gene




ID)


448
CSDC2; cold shock
CSDC2
PIPPIN;
NM_014460
ENSG00000172346
27254



domain containing C2,

dJ347H13.2



RNA binding;



ENSG00000172346


449
Gamma-parvin,
PARVG

NM_022141
ENSG00000138964
64098



ENSG00000138964,



PARVG


450
OTTHUMG00000030167,
CTA-
bK243E7.C22.3
No
OTTHUMG00000030167
No Available



CTA-243E7.3
243E7.3

Available




(Vega gene




ID)


451
Oncostatin M
OSM
MGC20461
NM_020530
ENSG00000099985
5008



precursor (OSM),



ENSG00000099985,



OSM


452
Oncostatin M
OSM
MGC20461
NM_020530
ENSG00000099985
5008



precursor (OSM),



ENSG00000099985,



OSM


453
Myosin-18B (Myosin
MYO18B
BK125H2.1
NM_014550
ENSG00000133454
84700



XVIIIb),



ENSG00000133454,



MYO18B


454
Q6ICL0_HUMAN
Q6ICL0_HUMAN
Em: AC006547.7,

ENSG00000184004
150197



(Predicted
(Predicted
FLJ32575



UniProt/TrEMBL ID),
UniProt/TrEMBL



hypothetical protein
ID)



FLJ3257;



ENSG00000184004


455
OTTHUMG00000030140,
CTA-
bA262A13.C22.5
No
OTTHUMG00000030140
No Available



CTA-299D3.6
299D3.6

Available




(Vega gene




ID)


456
GALR3; galanin
GALR3

NM_003614
ENSG00000128310
8484



receptor 3;



ENSG00000128310


457
GALR3; galanin
GALR3

NM_003614
ENSG00000128310
8484



receptor 3;



ENSG00000128310


458
IL2RB; interleukin 2
IL2RB
P70-75
NM_000878
ENSG00000100385
3560



receptor, beta;



ENSG00000100385


459
CTA-343C1.3 (Vega
CTA-
bK343C1.C22.3
No
OTTHUMG00000030151
No Available



gene ID); Putative
343C1.3

Available



Processed transcript;
(Vega gene



OTTHUMG00000030151
ID)


460
CTA-941F9.6
CTA-

No
OTTHUMG00000030231
No Available



(Vega_gene ID)
941F9.6

Available




(Vega_gene




ID)


461
CTA-941F9.6
CTA-

No
OTTHUMG00000030231
No Available



(Vega_gene ID)
941F9.6

Available




(Vega_gene




ID)


462
LL22NC03-121E8.1
LL22NC03-
cN121E8.C22.1
No
OTTHUMG00000030676
No Available



(Vega gene ID); Novel
121E8.1

Available



Protein coding;
(Vega gene



OTTHUMG00000030676
ID)


463
Cytohesin-4,
PSCD4
CYT4
NM_013385
ENSG00000100055
27128



ENSG00000100055,



PSCD4


464
RP4-754E20_A.4
RP4-
dJ754E20A.C22.4
No
OTTHUMG00000030716
No Available



(Vega gene ID);
754E20_A.4

Available



Putative Processed
(Vega



transcript;
gene ID)



OTTHUMG00000030716


465
PIB5PA;
PIB5PA
PIPP;
NM_001002837
ENSG00000185133
27124



phosphatidylinositol

INPP5;



(4,5) bisphosphate 5-

MGC129984



phosphatase, A;



ENSG00000185133;



embryonic marker


466
no gene associated


467
PLA2G3;
PLA2G3
GIII-
NM_015715
ENSG00000100078
50487



ENSG00000100078;

SPLA2



phospholipase A2,



group III


468
PLA2G3;
PLA2G3
GIII-
NM_015715
ENSG00000100078
50487



ENSG00000100078;

SPLA2



phospholipase A2,



group III


469
DGCR2; DiGeorge
DGCR2
IDD;
NM_005137
ENSG00000070413
9993



syndrome critical

LAN;



region gene 2;

DGS-C;



ENSG00000070413

SEZ-12;





KIAA0163;





DKFZp686I1730


470
TCN2; transcobalamin
TCN2
TC2;
NM_000355
ENSG00000185339
6948



II; macrocytic anemia;

D22S676;



ENSG00000185339

D22S750


471
IGLL1;
IGLL1
IGO; 14.1;
NM_020070
ENSG00000128322
3543



immunoglobulin

IGL1;



lambda-like

IGL5;



polypeptide 1;

IGLL;



ENSG00000128322

IGVPB;





CD179b;





VPREB2;





IGLJ14.1


472
RP1-29C18.7 (Vega
RP1-
dJ29C18.C22.7
No
OTTHUMG00000030424
No Available



gene ID); Novel
29C18.7

Available



Processed transcript;
(Vega gene



OTTHUMG00000030424
ID)


473
IGLC1;
IGLC1
IGLC;
NG_000002
ENSG00000100208
3537



immunoglobulin

MGC40381;



lambda constant 1

MGC40425;



(Mcg marker);

MGC88359;



ENSG00000100208

MGC88777;





MGC88779;





MGC104999;





DKFZp667J0810;





Constant





region of





lambda





light





chains;





immunoglobulin





lambda





constant





region 1





(Mcg





marker)


474
APOBEC3B;
APOBEC3B
ARP4;
NM_004900
ENSG00000179750
9582



apolipoprotein B

ARCD3;



mRNA editing

PHRBNL;



enzyme, catalytic

APOBEC1L;



polypeptide-like 3B;

FLJ21201;



ENSG00000179750

DJ742C19.2


475
CRYBB1; crystallin,
CRYBB1

NM_001887
ENSG00000100122
1414



beta B1;



ENSG00000100122


476
CRYBA4; crystallin,
CRYBA4

NM_001886
ENSG00000196431
1413



beta A4;



ENSG00000196431


477
sushi domain
SUSD2
BK65A6.2,
NM_019601
ENSG00000099994
56241



containing 2,

FLJ22778



ENSG00000099994,



SUSD2


478
sushi domain
SUSD2
BK65A6.2,
NM_019601
ENSG00000099994
56241



containing 2,

FLJ22778



ENSG00000099994,



SUSD2


479
OTTHUMG00000030870,
CTA-
bK503F6.C22.1
No
OTTHUMG00000030870
No Available



Putative Processed
503F6.1

Available



transcript, CTA-
(Vega gene



503F6.1
ID)


480
embryonic marker,
KB-
Em: AP000577.C22.3
No
OTTHUMG00000030800
No Available



OTTHUMG00000030800,
1323B2.3

Available



KB-1323B2.3
(Vega gene




ID)


481
no gene associated


482
IGLV1-44;
IGLV1-44
V1-16;
NG_000002
ENSG00000186751
28823



immunoglobulin

IGLV144



lambda variable 1-44;



ENSG00000186751


483
IGLV1-44;
IGLV1-44
V1-16;
NG_000002
ENSG00000186751
28823



immunoglobulin

IGLV144



lambda variable 1-44;



ENSG00000186751


484
OTTHUMG00000030922,
RP3-
Em: AC002378.C22.2
No
OTTHUMG00000030922
No Available



RP3-438O4.2
438O4.2

Available




(Vega_gene




ID)


485
OTTHUMG00000030922,
RP3-
Em: AC002378.C22.2
No
OTTHUMG00000030922
No Available



RP3-438O4.2
438O4.2

Available




(Vega_gene




ID)


486
APOL4;
APOL4
APOLIV;
NM_030643
ENSG00000100336
80832



apolipoprotein L, 4;

APOL-IV



ENSG00000100336


487
OTTHUMG00000030852,
RP4-
DJ756G23.1,
XM_377720
OTTHUMG00000030852
150356



RP4-
756G23.1
LOC150356
XP_377720



756G23.1, novel
(Vega gene



processed transcript
ID)


488
ENSG00000100399,
Q96E60_HUMAN
RP4-
XM_377720
ENSG00000100399
150356



Q96E60_HUMAN
(Predicted
756G23.1
XP_377720




UniProt/TrEMBL
hypothetical




ID)
protein





BC012882,





LOC150356


489
Neutrophil cytosol
NCF4
p40phox
NM_000631
ENSG00000100365
4689



factor 4 (NCF-4)



(Neutrophil NADPH



oxidase factor 4) (p40-



phox) (p40phox).,



ENSG00000100365,



NCF4


490
Neutrophil cytosol
NCF4
p40phox
NM_000631
ENSG00000100365
4689



factor 4 (NCF-4)



(Neutrophil NADPH



oxidase factor 4) (p40-



phox) (p40phox).,



ENSG00000100365,



NCF4


491
Somatostatin receptor
SSTR3

NM_001051
ENSG00000183473
6753



type 3 (SS3R) (SSR-



28), D



ENSG00000183473,



SSTR3


492
Somatostatin receptor
SSTR3

NM_001051
ENSG00000183473
6753



type 3 (SS3R) (SSR-



28), D



ENSG00000183473,



SSTR3


493
Bcl-2 interacting killer
BIK
NBK,
NM_001197
ENSG00000100290
638



(Apoptosis inducer

BBC1



NBK) (BP4) (BIP1).,



ENSG00000100290,



BIK


494
GAS2-like protein 1
GAS2L1
GAR22
NM_006478
ENSG00000185340
10634



(Growth arrest-specific



2-like 1) (GAS2-



related protein on



chromosome 22)



(GAR22 protein),



ENSG00000185340,



GAS2L1


495
RP3-355C18.2 (Vega
RP3-
dJ355C18.C22.2
No
OTTHUMG00000030072
No Available



gene ID)
355C18.2

Available




(Vega gene




ID)


496
SOX10; SRY (sex
SOX10
DOM;
NM_006941
ENSG00000100146
6663



determining region Y)-

WS4;



box 10;

MGC15649



ENSG00000100146


497
Gamma-parvin
PARVG

NM_022141
ENSG00000138964
64098



ENSG00000138964


498
Caspase recruitment
CARD10
CARMA3,
NM_014550
ENSG00000100065
29775



domain protein 10

BIMP1



(CARD-containing



MAGUK protein 3)



(Carma



3). ENSG00000100065,



CARD10


499
ENSG00000100101,
NP_077289.1
MGC3731;
NM_024313
ENSG00000100101
79159



NP_077289.1

dJ37E16.7


500
HTF9C; HpaII tiny
HTF9C_HUMAN
HTF9C;
NM_022727
ENSG00000099899
27037



fragments locus 9C;
(UniProt/Swiss-
MGC102728



ENSG00000099899
Prot




ID)


501
Oncostatin M
OSM
MGC20461
NM_020531
ENSG00000099985
5008



precursor (OSM),



ENSG00000099985,



OSM


502
CTA-407F11.4 (Vega
CTA-
bK407F11.C22.4
No
OTTHUMG00000030804
No Available



gene ID); Novel
407F11.4

Available



Processed transcript;
(Vega gene



OTTHUMG00000030804
ID)


503
Q6ICL0_HUMAN
Q6ICL0_HUMAN
Em: AC006547.7,

ENSG00000184004
150197



(Predicted
(Predicted
FLJ32575



UniProt/TrEMBL ID),
UniProt/TrEMBL



hypothetical protein
ID)



FLJ3257;



ENSG00000184004


504
CTA-989H11.2 (Vega
CTA-
bK989H11.C22.2
No
OTTHUMG00000030141
No Available



gene ID); Putative
989H11.2

Available



Processed transcript;
(Vega gene



OTTHUMG00000030141
ID)


505
transmembrane
TMPRSS6

NM_153609
ENSG00000187045
164656



protease, serine 6


506
HMG2L1; high-
HMG2L1
HMGBCG;
NM_001003681
ENSG00000100281
10042



mobility group protein

THC211630



2-like 1;



ENSG00000100281


507
NP_001017964.1
NP_001017964.1
LOC150223;
NM_001017964
ENSG00000161179
150223



(RefSeq peptide ID);
(RefSeq
MGC133160



hypothetical protein
peptide ID)



LOC150223;



ENSG00000161179


508
Platelet-derived
PDGFB
SIS; SSV;
NM_002608
ENSG00000100311
5155



growth factor B chain

PDGF2; c-



precursor (PDGF B-

sis



chain,



ENSG00000100311,



PDGFB


509
OTTHUMG00000030815,
CTA-
Em: U62317.C22.15
No
OTTHUMG00000030815
No Available



CTA-384D8.15
384D8.15

Available




(Vega gene




ID)


510
MGAT3; mannosyl
MGAT3
GNT3;
NM_002409
ENSG00000128268
4248



(beta-1,4-)-

GNT-III



glycoprotein beta-1,4-



N-



acetylglucosaminyltransferase;



ENSG00000128268


511
Ceramide kinase (EC
CERK
LK4;
NM_022766
ENSG00000100422
64781



2.7.1.138)

hCERK;



(Acylsphingosine

FLJ21430;



kinase) (hCERK)

FLJ23239;



(Lipid kinase 4)

KIAA1646;



(LK4),

MGC131878;



ENSG00000100422,

dA59H18.2;



CERK

dA59H18.3;





DKFZp434E0211


512
Reticulon 4 receptor
RTN4R
NGR;
NM_023004
ENSG00000040608
65078



precursor (Nogo

NOGOR



receptor) (NgR)



(Nogo-66 receptor),



ENSG00000040608,



RTN4R


513
UNC84B; unc-84
UNC84B
SUN2;
NM_015374
ENSG00000100242
25777



homolog B (C. Elegans);

KIAA0668



ENSG00000100242


514
RABL4; RAB,
RABL4
RAYL
NM_006860
ENSG00000100360
11020



member of RAS



oncogene family-like



4; ENSG00000100360


515
Cadherin EGF LAG
CELSR1
ME2,
NM_014246
ENSG00000075275
9620



seven-pass G-type

HFMI2,



receptor 1 precursor

FMI2,



(Flamingo homolog 2)

CDHF9



(hFmi2),



ENSG00000075275,



CELSR1


516
OTTHUMG00000030326,
LL22NC03-
cN5H6.C22.1
No
OTTHUMG00000030326
No Available



LL22NC03-
5H6.1

Available



5H6.1
(Vega gene




ID)


517
OTTHUMG00000030656,
RP3-
Em: AC005005.C22.6
No
OTTHUMG00000030656
No Available



RP3-515N1.6
515N1.6

Available




(Vega gene




ID)


518
SMTN; smoothelin;
SMTN

NM_006932
ENSG00000183963
6525



ENSG00000183963


519
ZNRF3 protein
ZNRF3
KIAA1133,
XM_290972,
ENSG00000183579
84133



(Fragment),

BK747E2.3,
XP_290972



ENSG00000183579,

FLJ22057,



ZNRF3 zinc and ring

RNF203



finger 3 (ZNRF3)


520
OTTHUMG00000030700,
GRAP2
Grf40,

OTTHUMG00000030700
9402



GRB2-related

GrbX,



adaptor protein 2,

GRBLG,



GRAP2

GADS,





Mona,





P38;





GRID;





GRPL;





GRB2L;





GRAP-2


521
CAP-binding protein
NP_073622.2
FLJ23588;
NM_022785
ENSG00000186976
64800



complex interacting
(RefSeq
DJBP;



protein 1 isoform a
peptide ID)
HSCBCIP1;



Source:

KIAA1672;



RefSeq_peptide

dJ185D5.1



NP_073622


522
SAM50_HUMAN
SAMM50
OMP85;
NM_015380
ENSG00000100347
25813



(UniProt/Swiss-Prot

SAM50;



ID),

TOB55;



ENSG00000100347,

TRG-3;



SAM50-like protein

CGI-51;



CGI-51; sorting and

YNL026W



assembly machinery



component 50



homolog (S. Cerevisiae)


523
SULT4A1;
SULT4A1
NST;
NM_014351
ENSG00000130540
25830



sulfotransferase family

BRSTL1;



4A, member 1;

SULTX3;



ENSG00000130540

BR-STL-1;





MGC40032;





DJ388M5.3;





hBR-





STL-1


524
TIMP3; TIMP
TIMP3
SFD;
NM_000362
ENSG00000100234
7078



metallopeptidase

K222;



inhibitor 3 (Sorsby

K222TA2;



fundus dystrophy,

HSMRK222



pseudoinflammatory);



ENSG00000100234


525
T-box transcription
TBX1
DGS;
NM_005992
ENSG00000184058
6899



factor TBX1 (T-box

TGA;



protein 1) (Testis-

CAFS;



specific T-box

CTHM;



protein),

DGCR;



ENSG00000184058,

DORV;



TBX1, TBX1 is

VCFS;



involved in heart

TBX1C



development-Great


526
ENSG00000186732,
MPPED1
239AB;
NM_001585
ENSG00000186732
758



metallophosphoesterase

FAM1A;



domain containing 1

C22orf1;



NM_001585.2

MGC88045


527
ENSG00000188511,
NP_942148.1
LOC348645
NM_198851
ENSG00000188511
348645



NP_942148.1 novel
(RefSeq



Gene hypothetical
peptide ID)



protein LOC348645


528
Cdc42 effector protein
CDC42EP1
MSE55,
NM_007061
ENSG00000128283
11135



1,

CEP1,



ENSG00000128283,

Borg5,



CDC42EP1

MGC15316


529
RPL3; ribosomal
RPL3
TARBP-B;
NM_000967
ENSG00000100316
6122



protein L3;

MGC104284



ENSG00000100316


530
APOL2;
APOL2
APOL-II
NM_030882
ENSG00000128335
23780



apolipoprotein L, 2;



ENSG00000128335


531
RAC2; ras-related C3
RAC2
Gx; EN-7;
NM_002872
ENSG00000128340
5880



botulinum toxin

HSPC022



substrate 2 (rho



family, small GTP



binding protein Rac2);



ENSG00000128340


532
OTTHUMP00000028917,
Q96E60_HUMAN
RP4-
XM_377720
ENSG00000100399
150356



ENSG00000100399,
(Predicted
756G23.1
XP_377720



Q96E60_HUMAN
UniProt/TrEMBL
hypothetical




ID)
protein





BC012882,





LOC150356


533
Neutrophil cytosol
NCF4
p40phox
NM_000631
ENSG00000100365
4689



factor 4 (NCF-4)



(Neutrophil NADPH



oxidase factor 4) (p40-



phox) (p40phox).,



ENSG00000100365,



NCF4


534
XP_371837.1 (RefSeq
XP_371837.1

No
ENSG00000168768
No Available



peptide predicted ID);
(RefSeq

Available



PREDICTED: similar
peptide



to oxidoreductase
predicted



UCPA Source:
ID)



RefSeq_peptide_predicted



XP_371837;



ENSG00000168768


535
triggering receptor
TREML2
FLJ13693,
NM_024807
ENSG00000112195
79865



expressed on myeloid

TLT2,



cells-like 2,

dJ238O23.1



ENSG00000112195,



TREML2


536
TREML1; triggering
TREML1
TLT1;
NM_178174
ENSG00000161911
340205



receptor expressed on

TLT-1;



myeloid cells-like 1;

PRO3438;



ENSG00000161911

GLTL1825;





MGC119173;





dJ238O23.3


537
ENSG00000178199,
ZC3H12D
FLJ46041;
XM_291154
ENSG00000178199
340152



Q6ZRW2_HUMAN;

C6orf95;
XP_291154



zinc finger CCCH-

dJ281H8.1



type containing 12D


538
AIM1; absent in
AIM1
ST4
NM_001624
ENSG00000112297
202



melanoma1;



ENSG00000112297


539
NKG2D ligand 4
RAET1E
LETAL,
NM_139165
ENSG00000164520
135250



precursor (NKG2D

bA350J20.7,



ligand 4) (NKG2DL4)

ULBP4;



(N2DL-4) (Retinoic

MGC125308;



acid early transcript

MGC125309;



1E) (Lymphocyte

bA350J20.7



effector toxicity



activation ligand)



(RAE-1-like transcript



4) (RL-4),



ENSG00000164520,



RAET1E


540
Disheveled associated
DAAM2
KIAA0381;
NM_015345
ENSG00000146122
23500



activator of

MGC90515;



morphogenesis 2,

dJ90A20A.1;



ENSG00000146122,

RP1-



DAAM2

278E11.1


541
RP11-535K1.1 (Vega
RP11-

No
OTTHUMG00000014660
No Available



gene ID); Putative
535K1.1

Available



Processed transcript;
(Vega gene



OTTHUMG00000014660
ID)


542
OTTHUMG00000015679;
RP3-
dJ509I19.3
No
OTTHUMG00000015679
No Available



Novel Protein
509I19.3

Available



coding; RP3-509I19.3
(Vega gene




ID)


543
RP11-503C24.1 (Vega
RP11-
bA503C24.1
No
OTTHUMG00000016040
No Available



gene ID); Putative
503C24.1

Available



Processed transcript;
(Vega gene



OTTHUMG00000016040
ID)


544
GABRR2; gamma-
GABRR2

NM_002043
ENSG00000111886
2570



aminobutyric acid



(GABA) receptor, rho



2; ENSG00000111886


545
ANKRD6; ankyrin
ANKRD6

NM_014942
ENSG00000135299
22881



repeat domain 6;



ENSG00000135299


546
TXLNB; taxilin beta;
TXLNB
MDP77;
NM_153235
ENSG00000164440
167838



ENSG00000164440

LST001;





C6orf198;





dJ522B19.2;





DKFZp451A175


547
TXLNB; taxilin beta;
TXLNB
MDP77;
NM_153235
ENSG00000164440
167838



ENSG00000164440

LST001;





C6orf198;





dJ522B19.2;





DKFZp451A175


548
RP5-899B16.2 (Vega
RP5-
dJ899B16.2
No
OTTHUMG00000015698
No Available



gene ID); Putative
899B16.2

Available



Processed transcript;
(Vega gene



OTTHUMG00000015698
ID)


549
Probable G-protein
GPR116
KPG_001;
NM_015234
ENSG00000069122
221395



coupled receptor 116

KIAA0758;



precursor,

DKFZp564O1923



ENSG00000069122,



GPR116


550
RP11-146I2.1 (Vega
RP11-
dJ190J20.1
No
OTTHUMG00000014290
No Available



gene ID); Novel
146I2.1

Available



Processed transcript;
(Vega gene



OTTHUMG00000014290
ID)


551
GPR115; G protein-
GPR115
PGR18;
NM_153838
ENSG00000153294
221393



coupled receptor 115;

FLJ38076



ENSG00000153294


552
GPR126; G protein-
GPR126
DREG;
NM_001032394
ENSG00000112414
57211



coupled receptor 126;

VIGR;



ENSG00000112414

PS1TP2



embryonic marker


553
RP1-60O19.1 (Vega
RP1-
dJ60O19.1
No
OTTHUMG00000015305
No Available



gene ID); Known
60O19.1

Available



Processed transcript;
(Vega gene



OTTHUMG00000015305
ID)


554
OTTHUMG00000015313,
SCML4
RP1-
NM_198081
OTTHUMG00000015313
256380



RP1-47M23.1

47M23.1



SCML4 sex comb on

(Vega_gene



midleg-like 4

ID)



(Drosophila) [Homo




sapiens]



555
OTTHUMG00006004170,
CRISP2
TPX1;
NM_003296
OTTHUMG00000014822
7180



TPX1testis

TSP1;



specific protein 1

GAPDL5;



(probe H4-1 p3-1)

CRISP-2;





MGC111136


556
OTTHUMG00000014829,
RP11-
bA397G17.1
No
OTTHUMG00000014829
No Available



RP11-397017.1, novel
397G17.1

Available



processed transcript.
(Vega gene




ID)


557
OTTHUMG00000015337RP11-
RP11-
LOC389422
XM_374179
OTTHUMG00000015337
389422



487F23.3
487F23.3

XP_374179



hypothetical
(Vega gene



LOC389422
ID)


558
Nesprin-1 (Nuclear
SYNE1
SYNE-1B,
NM_015293
ENSG00000131018
23345



envelope spectrin

KIAA0796,



repeat protein 1)

8B,



(Synaptic nuclear

nesprin-1,



envelope protein 1)

enaptin,



(Syne-1) (Myocyte

MYNE1,



nuclear envelope

CPG2



protein 1) (Myne-1)



(Enaptin),



ENSG00000131018,



SYNE1


559
Nesprin-1 (Nuclear
SYNE1
SYNE-1B,
NM_015293
ENSG00000131018
23345



envelope spectrin

KIAA0796,



repeat protein 1)

8B,



(Synaptic nuclear

nesprin-1,



envelope protein 1)

enaptin,



(Syne-1) (Myocyte

MYNE1,



nuclear envelope

CPG2



protein 1) (Myne-1)



(Enaptin),



ENSG00000131018,



SYNE1


560
RP11-398K22.4 (Vega
RP11-
Em: AC019205.4
No
OTTHUMG0000001504
No Available



gene ID); Putative
398K22.4

Available



Processed transcript;
(Vega gene



OTTHUMG00000015024
ID)


561
MyoD family inhibitor
MDFI
I-MF
NM_005586
ENSG00000112559
4188



(Myogenic repressor I-



mf),



ENSG00000112559,



MDFI


562
OTTHUMG00000014691,
RP11-
bA533O20.2
No
OTTHUMG00000014691
No Available



putative
533O20.2

Available



processed transcript,
(Vega gene



RP11-533O20.2
ID)


563
RP3-398D13.4 (Vega
RP3-
dJ398D13.4
No
OTTHUMG00000014188
No Available



gene ID);
398D13.4

Available



OTTHUMG00000014188
(Vega gene




ID);


564
RP3-429O6.1 (Vega
RP3-
dJ429O6.1
No
OTTHUMG00000014195
No Available



gene ID); Putative
429O6.1

Available



Processed transcript;
(Vega gene



OTTHUMG00000014195
ID)


565
MOG; myelin
MOG
MGC26137
NM_001008228
ENSG00000137345
4340



oligodendrocyte



glycoprotein;



ENSG00000137345


566
RP3-495K2.2 (Vega
RP3-
dJ495K2.2
No
OTTHUMG00000016052
No Available



gene ID); Putative
495K2.2

Available



Processed transcript;
(Vega gene



OTTHUMG00000016052
ID)


567
RP11-417E7.1 (Vega
RP11-
bA417E7.1
No
OTTHUMG00000016054
No Available



gene ID); Putative
417E7.1

Available



Processed transcript;
(Vega gene



OTTHUMG00000016054
ID)


568
tyrosine-protein
PTK7
CCK4
NM_002821
ENSG00000112655
5754



kinase-like 7 precursor



(Colon carcinoma



kinase 4) (CCK-4).,



ENSG00000112655,



PTK7


569
RP11-174C7.4 (Vega
RP11-
bA174C7.4
No
OTTHUMG00000015553
No Available



gene ID)
174C7.4

Available




(Vega gene




ID)


570
cytidine
CMAH
CSAH;
NR_002174
OTTHUMG00000016099
8418



monophosphate-N-

CMP-



acetylneuraminic acid

NeuAc



hydroxylase (CMP-N-

hydroxylase;



acetylneuraminate

CMP-



monooxygenase);

Neu5Ac



CMAH

hydroxylase;





CMP-





sialic acid





hydroxylase;





CMP-N-





acetylneuraminic





acid





hydroxylase;





cytidine





monophos





pho-N-





acetylneuraminic





acid





hydroxylase


571
KHD1; polycystic
PKHD1
FCYT;
NM_138694
ENSG00000170927
5314



kidney and hepatic

ARPKD;



disease 1 (autosomal

TIGM1



recessive);



ENSG00000170927


572
RP3-471C18.2 (Vega
RP3-
dJ471C18.2
No
OTTHUMG00000014332
No Available



gene ID); Novel
471C18.2

Available



Processed transcript;
(Vega gene



OTTHUMG00000014332
ID)


573
RP11-204E9.1 (Vega
RP11-
bA204E9.1
No
OTTHUMG00000014342
No Available



gene ID); Putative
204E9.1

Available



Processed transcript;
(Vega gene



OTTHUMG00000014342
ID)


574
glutathione peroxidase
GPX5

NM_001509
OTTHUMG00000016307
2880



5,



OTTHUMG00000016307,



GPX5


575
RP11-411K7.1 (Vega
RP11-
bA411K7.1
No
OTTHUMG00000014887
No Available



gene ID); Putative
411K7.1

Available



Processed transcript;
(Vega gene



OTTHUMG00000014887
ID)


576
skin marker,
GRIK2
RP3-
NM_021956
ENSG00000164418
2898



Glutamate receptor,

438O4.2



ionotropic kainate 2

(Vega gene



precursor (Glutamate

ID)



receptor 6) (GluR-6)



(GluR6) (Excitatory



amino acid receptor 4)



(EAA4)


577
C6orf142;
C6orf142
MGC18257
NM_138569
ENSG00000146147
90523



chromosome 6 open



reading frame 142;



ENSG00000146147


578
HDGFL1; hepatoma
HDGFL1
PWWP1;
NM_138574
ENSG00000112273
154150



derived growth factor-

dJ309H15.1



like 1;



ENSG00000112273


579
forkhead box C1,
FOXC1
FKHL7,
NM_001453
OTTHUMG00000016182
2296



OTTHUMG00000016182,

IRID1,



FOXC1

FREAC3,





ARA,





IGDA,





IHG1


580
C6orf188;
C6orf188
MGC45451;
NM_153711
ENSG00000178033
254228



chromosome 6 open

dJ493F7.3



reading frame 188;



ENSG00000178033


581
ME1; malic enzyme 1,
ME1
MES;
NM_002395
ENSG00000065833
4199



NADP(+)-dependent,

HUMNDME



cytosolic;



ENSG00000065833


582
SLC22A1; solute
SLC22A1
OCT1;
NM_003057
ENSG00000175003
6580



carrier family 22



(organic cation



transporter), member 1


583
RP11-235G24.1 (Vega
RP11-
bA235G24.1
No
OTTHUMG00000015959
No Available



gene ID)
235G24.1

Available




(Vega gene




ID)


584
T-box 18; TBX18
TBX18

XM_496819
ENSG00000112837
9096






XP_496819


585
CTA-31J9.2, putative
CTA-
bK31J9.2
No
OTTHUMG00000015619
No Available



processed transcript,
31J9.2

Available



OTTHUMG00000015619
(Vega gene




ID)


586
RP1-32B1.4 (Vega
RP1-
dJ32B1.4
No
OTTHUMG00000015628
No Available



gene ID); Putative
32B1.4

Available



Processed transcript
(Vega gene



OTTHUMG00000015628
ID)


587
OTTHUMG00000014223,
RP11-
bA203H2.2
No
OTTHUMG00000014223
No Available



RP11-203H2.2,
203H2.2

Available



novel processed
(Vega gene



treanscript
ID)


588
OTTHUMG00000014737,
C6orf154
MGC131686;
NM_001012974
OTTHUMG00000014737
221424



C6orf154 and

dJ337H4.2



Name: chromosome 6



open reading frame



154; RP3-337H4.2


589
transcription factor
TFAP2A
AP-2
NM_001032280
OTTHUMG00000014235
7020



AP-2 alpha,



OTTHUMG00000014235,



TFAP2A


590
IL20RA; interleukin
IL20RA
IL-20R1;
NM_014432
ENSG00000016402
53832



20 receptor, alpha;

ZCYTOR7



ENSG00000016402


591
KAAG1; kidney
KAAG1
RU2;
NM_181337
ENSG00000146049
353219



associated antigen 1;

RU2AS;



ENSG00000146049

MGC78738


592
TGM3;
TGM3
TGE;
NM_003245
ENSG00000125780
7053



transglutaminase 3 (E

MGC126249;



polypeptide, protein-

MGC126250



glutamine-gamma-



glutamyltransferase);



ENSG00000125780


593
RASSF2; Ras
RASSF2
KIAA0168;
NM_014737
ENSG00000101265
9770



association

DKFZp781O1747



(RalGDS/AF-6)



domain family 2;



ENSG00000101265


594
no gene associated


595
no gene associated


596
no gene associated


597
no gene associated


598
no gene associated


599
no gene associated


600
no gene associated


601
no gene associated


602
no gene associated


603
no gene associated


604
no gene associated


605
RP4-697P8.2 (Vega
RP4-
dJ697P8.2
No
OTTHUMG00000031879
No Available



gene ID); Putative
697P8.2

Available



Processed transcript;
(Vega gene



OTTHUMG00000031879
ID)


606
no gene associated


607
OTTHUMG00000031883,
RP4-
DJ734C18.1
No
OTTHUMG00000031883
No Available



RP4-734C18.1,
734C18.1

Available



putative processed
(Vega gene



transcript
ID)


608
no gene associated


609
no gene associated


610
no gene associated


611
no gene associated


612
no gene associated


613
Ras and Rab interactor
RIN2
RASSF4
NM_018993
OTTHUMG00000031996
54453



2,



OTTHUMG00000031996,



RIN2


614
no gene associated


615
no gene associated


616
no gene associated


617
no gene associated


618
no gene associated


619
no gene associated


620
no gene associated


621
no gene associated


622
no gene associated


623
no gene associated


624
C20orf112;
C20orf112
dJ1184F4.2;
NM_080616
OTTHUMG00000032219
140688



chromosome 20 open

DKFZP566G1424



reading frame 112;



OTTHUMG00000032219


625
FER1L4; fer-1-like 4
FER1L4
FLJ13459;
NR_001442
OTTHUMG00000032354
80307



(C. Elegans);

FLJ22613;



OTTHUMG00000032354

C20orf124;





dJ309K20.1;





bA563A22B.1;





fer-1





(C. elegans)-





like 4


626
no gene associated


627
no gene associated


628
Protein C20orf102



precursor,



ENSG00000132821,



CT102_HUMAN


629
no gene associated


630
no gene associated


631
no gene associated


632
no gene associated


633
no gene associated -



Nearest transcript



CDH22 (~18 kb



upstream)


634
no gene associated


635
no gene associated


636
no gene associated


637
no gene associated


638
no gene associated


639
no gene associated


640
no gene associated


641
ZHX3; zinc fingers
ZHX3
TIX1;
NM_015035
OTTHUMG00000032481
23051



and homeoboxes 3;

KIAA0395



OTTHUMG00000032481


642
no gene associated


643
CHD6; chromodomain
CHD6
CHD5;
NM_032221
ENSG00000124177
84181



helicase DNA binding

RIGB;



protein 6;

KIAA1335



ENSG00000124177


644
no gene associated


645
PTPRG; protein
PTPRG
PTPG;
NM_002841
ENSG00000144724
5793



tyrosine phosphatase,

HPTPG;



receptor type, G;

RPTPG;



ENSG00000144724

R-PTP-



Gene is located on

GAMMA



Chr. 3


646
no gene associated


647
no gene associated


648
no gene associated


649
PTPNS1; protein
PTPNS1
BIT; MFR;
NM_080792
ENSG00000198053
140885



tyrosine phosphatase,

P84; SIRP;



non-receptor type

MYD-1;



substrate 1;

SHPS1;



ENSG00000198053

SHPS-1;





SIRPalpha;





SIRPalpha





2; SIRP-





ALPHA-1


650
Q7Z5T1_HUMAN
Q7Z5T1_HUMAN
KIAA1442;
XM_044921
ENSG00000088881
57593



(Predicted
(Predicted
EBF4;
XP_044921



UniProt/TrEMBL ID);
UniProt/TrEMBL
O/E-4;



KIAA1442 protein;
ID)
RP5-



ENSG00000088881

860F19.3


651
NP_689717.2 (RefSeq
NP_689717.2

No
ENSG00000171984
No Available



peptide ID);
(RefSeq

Available



ENSG00000171984
peptide ID)


652
ENSG00000149346,
C20orf94
NP_001009608.1,
NM_001009608
ENSG00000149346
128710



NP_001009608.1,

bA204H22.1;



hypothetical protein

bA254M13.1;



LOC128710,

dJ1099D15.3



chromosome 20 open



reading frame 94


653
C20orf82;
C20orf82
bA149I18.1;
XM_097736
ENSG00000101230
140862



chromosome 20 open

dJ1077I2.1
XP_097736



reading frame 82;



ENSG00000101230


654
C20orf23;
C20orf23
SNX23;
NM_024704
ENSG00000089177
55614



chromosome 20 open

KISC20ORF



reading frame 23;



ENSG00000089177;



embryonic marker


655
PCSK2; proprotein
PCSK2
PC2;
NM_002594
ENSG00000125851
5126



convertase

NEC2;



subtilisin/kexin type 2;

SPC2



ENSG00000125851


656
PCSK2; proprotein
PCSK2
PC2;
NM_002594
ENSG00000125851
5126



convertase

NEC2;



subtilisin/kexin type 2;

SPC2



ENSG00000125851


657
solute carrier family
SLC24A3
NCKX3
NM_020689
OTTHUMG00000031993
57419



24



(sodiumVpotassiumVcalcium



exchanger),



member 3,



OTTHUMG00000031993,



SLC24A3 (


658
solute carrier family
SLC24A3
NCKX3
NM_020689
OTTHUMG00000031993
57419



24



(sodiumVpotassiumVcalcium



exchanger),



member 3,



OTTHUMG00000031993,



SLC24A3 (


659
ENSG00000089101,
C20orf26
dJ1178H5.4;
NM_015585
ENSG00000089101
26074



CT026_HUMAN

DKFZP434K156


660
ENSG00000089101,
C20orf26
dJ1178H5.4;
NM_015585
ENSG00000089101
26074



CT026_HUMAN

DKFZP434K156


661
C20orf74 protein,
Q9ULE8_HUMAN
FLJ12819;
XM_046600
ENSG00000188559
57186



ENSG00000188559,
(Predicted
KIAA1272;
XP_046600



Q9ULE8_HUMAN
UniProt/TrEMBL
bA287B20.1;




ID)
dJ1049G11.4;





RP11-





470C13.2


662
C20orf74 protein,
Q9ULE8_HUMAN
FLJ12819;
XM_046600
ENSG00000188559
57186



ENSG00000188559,
(Predicted
KIAA1272;
XP_046600



Q9ULE8_HUMAN
UniProt/TrEMBL
bA287B20.1;




ID)
dJ1049G11.4;





RP11-





470C13.2


663
C20orf74 protein,
Q9ULE8_HUMAN
FLJ12819;
XM_046600
ENSG00000188559
57186



ENSG00000188559,
(Predicted
KIAA1272;
XP_046600



Q9ULE8_HUMAN
UniProt/TrEMBL
bA287B20.1;




ID)
dJ1049G11.4;





RP11-





470C13.2


664
PLAGL2;
PLAGL2

NM_002657
ENSG00000126003
5326



pleiomorphic adenoma



gene-like 2;



ENSG00000126003


665
GGTL3; gamma-
GGTL3
GGTL5;
NM_052830
ENSG00000131067
2686



glutamyltransferase-

D20S101;



like 3;

dJ18C9.2



ENSG00000131067


666
MYH7B; myosin,
MYH7B
MYH14;
NM_020884
ENSG00000078814
57644



heavy polypeptide 7B,

MGC96928;



cardiac muscle, beta;

MGC96940



ENSG00000078814


667
TRPC4AP; transient
TRPC4AP
TRUSS;
NM_015638
ENSG00000100991
26133



receptor potential

TRRP4AP;



cation channel,

C20orf188



subfamily C, member



4 associated protein;



ENSG00000100991


668
EPB41L1; erythrocyte
EPB41L1
4.1N;
NM_012156
ENSG00000088367
2036



membrane protein

KIAA0338;



band 4.1-like 1;

MGC11072



ENSG00000088367


669
C20orf117;
C20orf117
FLJ44670;
NM_080627
OTTHUMG00000032395
140710



chromosome 20 open

KIAA0889;



reading frame 117;

dJ132F21.1



OTTHUMG00000032395


670
PTPRT; protein
PTPRT
RPTPrho;
NM_007050
ENSG00000196090
11122



tyrosine phosphatase,

KIAA0283



receptor type, T;



ENSG00000196090


671
PTPRT; protein
PTPRT
RPTPrho;
NM_007050
ENSG00000196090
11122



tyrosine phosphatase,

KIAA0283



receptor type, T;



ENSG00000196090


672
PTPRT; protein
PTPRT
RPTPrho;
NM_007050
ENSG00000196090
11122



tyrosine phosphatase,

KIAA0283



receptor type, T;



ENSG00000196090


673
PTPRT; protein
PTPRT
RPTPrho;
NM_007050
ENSG00000196090
11122



tyrosine phosphatase,

KIAA0283



receptor type, T;



ENSG00000196090


674
PTPRT; protein
PTPRT
RPTPrho;
NM_007050
ENSG00000196090
11122



tyrosine phosphatase,

KIAA0283



receptor type, T;



ENSG00000196090


675
SDC4; syndecan 4
SDC4
SYND4;
NM_002999
ENSG00000124145
6385



(amphiglycan,

MGC22217



ryudocan);



ENSG00000124145


676
SDC4; syndecan 4
SDC4
SYND4;
NM_002999
ENSG00000124145
6385



(amphiglycan,

MGC22217



ryudocan);



ENSG00000124145


677
cadherin-like 22,
CDH22
C20orf25;
NM_021248
OTTHUMG00000033073
64405



OTTHUMG00000033073,

dJ998H6.1;



CDH22

MGC39564


678
EYA2; eyes absent
EYA2
EAB1;
NM_005244
ENSG00000064655
2139



homolog 2

MGC10614



(Drosophila);



ENSG00000064655


679
SULF2; sulfatase 2;
SULF2
HSULF-2;
NM_018837
ENSG00000196562
55959



ENSG00000196562

KIAA1247;





MGC126411;





DKFZp313E091;





RP5-





1049G16.1


680
KCNB1; potassium
KCNB1
DRK1;
NM_004975
ENSG00000158445
3745



voltage-gated channel,

KV2.1; h-



Shab-related

DRK1



subfamily, member 1;



ENSG00000158445


681
Breast carcinoma
BCAS4
FLJ20495,
NM_001010974
ENSG00000124243
55653



amplified sequence 4,

BHLHB4



ENSG00000124243,



BCAS4


682
nuclear factor of
NFATC2
NF-ATP,

OTTHUMG00000032747
4773



activated T-cells,

NFATp



cytoplasmic,



calcineurin-dependent



2,



OTTHUMG00000032747,



NFATC2


683
Nuclear factor of
NFATC2
NF-ATP,
NM_012340
ENSG00000101096
4773



activated T-cells,

NFATp



cytoplasmic 2 (T cell



transcription factor



NFAT1) (NFAT pre-



existing subunit) (NF-



ATp),



ENSG00000101096,



NFATC2


684
Bone morphogenetic
BMP7
OP-1
NM_001719
ENSG00000101144
655



protein 7 precursor



(BMP-7) (Osteogenic



protein 1) (OP-1)



(Eptotermin alfa),



ENSG00000101144,



BMP7


685
transmembrane,
TMEPAI
STAG1;
NM_020182
OTTHUMG00000032831
56937



prostate androgen

PMEPA1



induced RNA,



OTTHUMG00000032831,



TMEPAI


686
ENSG00000176659,
NP_775915.1

No
ENSG00000176659
No Available



NP_775915.1
(RefSeq

Available




peptide ID)


687
CDH4; cadherin 4,
CDH4
CAD4;
NM_001794
ENSG00000179242
1002



type 1, R-cadherin

RCAD;



(retinal);

FLJ22202;



ENSG00000179242

FLJ40547;





MGC126700


688
NP_001002034.1
NP_001002034.1

No
ENSG00000177096
No Available



(RefSeq peptide ID);
(RefSeq

Available



ENSG00000177096
peptide ID)


689
NP_612444.1 (RefSeq
NP_612444.1

No
ENSG00000133477
No Available



peptide ID);
(RefSeq

Available



ENSG00000133477
peptide ID)


690
no gene associated


691
OTTHUMG00000030780,
CTA-
bK373H7.C22.4
No
OTTHUMG00000030780
No Available



CTA-373H7.4, novel
373H7.4

Available



pseudogene
(Vega gene




ID)


692
no gene associated


693
Cat eye syndrome
CECR1
IDGFL
NM_017424
ENSG00000093072
51816



critical region protein



1 precursor,



ENSG00000093072,



CECR1


694
IGLC1;
IGLC1
IGLC;
NG_000002
ENSG00000100208
3537



immunoglobulin

MGC40381;



lambda constant 1

MGC40425;



(Mcg marker);

MGC88359;



ENSG00000100208

MGC88777;





MGC88779;





MGC104999;





DKFZp667J0810;





Constant





region of





lambda





light





chains;





immunoglobulin





lambda





constant





region 1





(Mcg





marker)


695
OTTHUMG00000030521,
AC000095.4
Em: AC000095.C22.4
No
OTTHUMG00000030521
No Available



AC000095.4
(Vega

Available



putative processed
gene ID)



transcript;


696
Uroplakin-3A
UPK3A
UPK3;
NM_006953
ENSG00000100373
7380



precursor (Uroplakin

UPIII



III) (UPIII).,



ENSG00000100373,



UPK3A


697
Spl site_no gene



associated


698
USP18; ubiquitin
USP18
ISG43;
NM_017414
OTTHUMG00000030949
11274



specific peptidase 18;

UBP43



OTTHUMG00000030949


699
BCR; breakpoint
BCR
ALL;
NM_004327
ENSG00000186716
613



cluster region;

CML;



ENSG00000186716

PHL;





BCR1;





D22S11;





D22S662


700
TBC1D10A; TBC1
TBC1D10A
EPI64;
NM_031937
ENSG00000099992
83874



domain family,

TBC1D10;



member 10A;

dJ130H16.1;



ENSG00000099992

AC004997.C22.2


701
signal peptide-CUB
SCUBE1

NM_173050
ENSG00000159307
80274



domian-EGF-related 1,



ENSG00000159307,



SCUBE1


702
MAPK8IP2; mitogen-
MAPK8IP2
IB2; JIP2;
NM_012324
ENSG00000008735
23542



activated protein

PRKM8IPL



kinase 8 interacting



protein 2;



ENSG00000008735


703
ENSG00000192797,
No

NM_000407
ENSG00000192797
No Available



miRNA
Available


704
RPL3; ribosomal
RPL3
TARBP-B;
NM_000967
ENSG00000100316
6122



protein L3;

MGC104284



ENSG00000100316


705
RPL3; ribosomal
RPL3
TARBP-B;
NM_000967
ENSG00000100316
6122



protein L3;

MGC104284



ENSG00000100316


706
RP4-695O20_B.9
RP4-
dJ695O20B.C22.9
No
OTTHUMG00000030111
No Available



(Vega gene ID);
695O20_B.9

Available



Putative Processed
(Vega



transcript;
gene ID)



OTTHUMG00000030111


707
NOVEL transcript??



No associated gene


708
MN1; meningioma
MN1
MGCR;
NM_002430
ENSG00000169184
4330



(disrupted in balanced

MGCR1;



translocation) 1;

MGCR1-



ENSG00000169184

PEN;





dJ353E16.2


709
no gene associated


710
RTDR1; rhabdoid
RTDR1
MGC16968
NM_014433
ENSG00000100218
27156



tumor deletion region



gene 1;



ENSG00000100218


711
RPL3; ribosomal
RPL3
TARBP-B;
NM_000967
ENSG00000100316
6122



protein L3;

MGC104284



ENSG00000100316


712
embryonic marker,
GRAP2
Grf40,

OTTHUMG00000030700
9402



GRB2-related adaptor

GrbX,



protein 2,

GRBLG,



OTTHUMG00000030700,

GADS,



GRAP2

Mona,





P38;





GRID;





GRPL;





GRB2L;





GRAP-2


713
Serine/threonine-
STK19
G11; RP1;
NM_032454
ENSG00000166301
8859



protein kinase 19 (EC

D6S60;



2.7.1.37) (RP1 protein)

D6S60E;



(G11 protein).

HLA-RP1;





MGC117388


714
Transcription factor 19
TCF19_HUMAN
SC1
Q9Y242
ENSG00000137310



(Transcription factor



SC1).


715
Pannexin-2
PANX2
hPANX2;
NM_052839
ENSG00000073150
56666





MGC119432


716

CTA-
bK243E7.C22.3

OTTHUMG00000030167




243E7.3


717
signal peptide-CUB
SCUBE1

NM_173050
ENSG00000159307
80274



domian-EGF-related 1


718
Reticulon 4 receptor
RTN4R
NGR;
NM_023004
ENSG00000040608
65078



precursor (Nogo

NOGOR



receptor) (NgR)



(Nogo-66 receptor)


719
Arylsulfatase A
ARSA
MLD
NM_000487
ENSG00000100299
410



precursor (EC 3.1.6.8)



(ASA) (Cerebroside-



sulfatase) [Contains:



Arylsulfatase A



component B;



Arylsulfatase A



component C]


720
glycoprotein Ib
GP1BB
CD42c
NM_000407
OTTHUMG00000030191
2812



(platelet), beta



polypeptide


721


722

No gene




associated


723
Mitochondrial
SLC25A18
GC2
NM_031481
ENSG00000182902
83733



glutamate carrier 2



(Glutamate/H(+)



symporter 2) (Solute



carrier family 25



member 18,



ENSG00000182902,



SLC25A18


724
Thioredoxin reductase
TXNRD2
TR; TR3;
NM_006440.2
ENSG00000184470
10587



2, mitochondrial

SELZ;



precursor (EC 1.8.1.9)

TRXR2;



(TR3) (TR-beta)

TR-BETA



(Selenoprotein Z)



(SelZ)


725
Somatostatin receptor
SSTR3

NM_001051
ENSG00000183473
6753



type 3 (SS3R) (SSR-



28)


726

RP11-
bA191L9.C22.1

OTTHUMG00000030964




191L9.1


727
No description-
AP000357.3
Em: AP000357.C22.3

OTTHUMG00000030574



pseudogene


728
Cat eye syndrome
CECR1
IDGFL
NM_017424.2
ENST00000262607
51816



critical region protein



1 precursor


729

No gene




associated


730
Membrane protein
MLC1
VL; LVM;
NM_015166
ENSG00000100427
23209



MLC1

MLC;





KIAA0027


731
BAI1-associated
BAIAP2L2
FLJ22582
NM_025045.3
ENSG00000128298
80115



protein 2-like 2


732
No description
NP_056185.1


ENSG00000100249


733
No description
RP4-
dJ695O20B.C22.9

OTTHUMG00000030111




695O20_B.9


734
OTTHUMG00000030167,
CTA-
bK243E7.C22.3
No
OTTHUMG00000030167
No Available



CTA-243E7.3
243E7.3

Available




(Vega gene




ID)


735
novel transcript
XXbac-
Em: AC006547.C22.7

OTTHUMG00000030620




B444P24.7


736

LL22NC03-


OTTHUMG00000030676




121E8.1-




001


737
No description
Q6ZN90_HUMAN


ENSG00000197549


738
NFAT activation
NFAM1
CNAIP;
NM_145912
ENSG00000167087
150372



molecule 1 precursor

FLJ40652



(Calcineurin/NFAT-



activating ITAM-



containing protein)



(NFAT activating



protein with ITAM



motif 1).


739
immunoglobulin
IGLC2
IGLC;

OTTHUMG00000030352
3538



lambda constant 2

MGC20392;





MGC45681;





Ig light-





chain,





partial Ke-





Oz-





polypeptide,





C-term;





immunoglobulin





lambda





constant





region 2





(Kern-Oz-





marker)


740
immunoglobulin
IGLC2
IGLC;

OTTHUMG00000030352
3538



lambda constant 2

MGC20392;





MGC45681;





Ig light-





chain,





partial Ke-





Oz-





polypeptide,





C-term;





immunoglobulin





lambda





constant





region 2





(Kern-Oz-





marker)


741
OTTHUMG00000030870,
CTA-
bK503F6.C22.1
No
OTTHUMG00000030870
No Available



CTA-503F6.1
503F6.1

Available




(Vega_gene




ID)


742
Lactosylceramide 4-
A4GALT
P1; PK;
NM_017436
ENSG00000128274
53947



alpha-

A14GALT;



galactosyltransferase

A4GALT1



(EC 2.4.1.228)


743

RP11-


OTTHUMG00000030966




191L9.3


744
Cold shock domain
CSDC2_HUMAN
PIPPIN
Q9Y534
ENSG00000172346



protein C2 (RNA-



binding protein



PIPPin)


745

GAS2L1
GAR22;
NM_152236.1
ENSG00000185340
10634





MGC17243


746
BAI1-associated
BAIAP2L2
FLJ22582
NM_025045.3
ENSG00000128298
80115



protein 2-like 2


747

NP_997360.1


ENSG00000197182


748
OTTHUMG00000030991,
LL22NC03-
dJ671O14.C22.6;
No
OTTHUMG00000030991
No Available



LL22NC03-
75B3.6
KIAA1644
Available



75B3.6
(Vega gene




ID)


749
Reticulon 4 receptor
RTN4R
NGR;
NM_023004
ENSG00000040608
65078



precursor (Nogo

NOGOR



receptor) (NgR)



(Nogo-66 receptor)


750
Smoothelin
SMTN

NM_134269
ENSG00000183963
6525


751
solute carrier family
SLC35E4

NM_001001479.1
ENSG00000100036
339665



35, member E4


752
protein C22orf13
CV013_HUMAN

Q96NT3
ENSG00000138867



(Protein LLN4)


753

No gene




associated


754
Histone
HIST1H3A
H3/A;
NM_003532.2
ENSG00000196966
8350





H3FA


755
Gamma-aminobutyric-
GABRR1 (

NM_002042
ENSG00000146276
2569



acid receptor rho-1



subunit precursor



(GABA(A) receptor).


756
OTTHUMG00000015693,
RP11-
bA12A2.3
No
OTTHUMG00000015693
No Available



RP11-12A2.3
12A2.3

Available




(Vega_gene




ID)


757

RP5-
dJ899B16.1

OTTHUMG00000015697




899B16.1 (


758
No description
RP11-
bA146I2.2

OTTHUMG00000014289




146I2.2


759

NP_060483.2


ENSG00000178289


760
Forkhead box protein
FOXO3A
AF6q21;
NM_001455
ENSG00000118689
2309



O3A,

FKHRL1;



ENSG00000118689,

FKHRL1P2;



FOXO3A

MGC12739;





MGC31925;





DKFZp781A0677


761
nuclear receptor
NCOA7
ESNA1;
XM_059748
ENSG00000111912
135112



coactivator 7

ERAP140;





MGC88425;





Nbla00052;





Nbla10993;





dJ187J11.3


762

RP11-
bA554D15.1

OTTHUMG00000015043




554D15.1


763
chromosome 6 open
C6orf190
C6orf207;
XM_069189
OTTHUMG00000015534
387357



reading frame 190

FLJ40584;





bA325O24.3;





bA325O24.4


764
phosphatase and actin
PHACTR2
C6orf56;
NM_014721
OTTHUMG00000015732
9749



regulator 2

KIAA0680;





DKFZp686F18175


765
High mobility group
HMGA1
HMG-R;
NM_002131
ENSG00000137309
3159



protein HMG-I/HMG-

HMGIY;



Y (HMG-I(Y)) (High

MGC4242;



mobility group AT-

MGC4854;



hook 1) (High mobility

MGC12816



group protein A1),



ENSG00000137309,



HMGA1


766
Pantetheinase
VNN1
Tiff66;
NM_004666
ENSG00000112299
8876



precursor (EC 3.5.1.—),

MGC116930;



ENSG00000112299,

MGC116931;



VNN1

MGC116932;





MGC116933


767
histone H2A
HIST1H2AA
H2AFR;
NM_170745
ENSG00000164508
221613





bA317E16.2


768
transcription factor
TFAP2A
AP-2;
NM_003220
OTTHUMG00000014235
7020



AP-2 alpha (activating

AP2TF;



enhancer binding

TFAP2;



protein 2 alpha)

AP-2alpha


769
N-acetyllactosaminide
GCNT2
II,
NM_001491
ENSG00000111846
2651



beta-1,6-N-

GCNT5;



acetylglucosaminyl-

II; IGNT;



tansferase (EC

ULG3;



2.4.1.150),

AIGnT;



ENSG00000111846,

BIGnT;



GCNT2

CIGnT;





GCNT5;





NAGCT1;





bA421M1.1;





bA360O19.2


770

No gene




associated


771

No gene




associated


772

No gene




associated


773

No gene




associated


774

No gene




associated


775

No gene




associated


776

No gene




associated


777

No gene




associated


778

No gene




associated


779

No gene




associated


780
No description
RP11-
bA318C17.1

OTTHUMG00000031920




318C17.1


781

No gene




associated


782

No gene




associated


783

No gene




associated


784

No gene




associated


785

No gene




associated


786

No gene




associated


787
novel transcript
RP11-


OTTHUMG00000032045




216C10.1


788

No gene




associated


789

No gene




associated


790

No gene




associated


791

RP11-
bA410N8.3

OTTHUMG00000032221




410N8.3


792

TIMP3 (
SFD;
NM_000362
ENSG00000100234
7078





K222;





K222TA2;





HSMRK222


793

No gene




associated


794

No gene




associated


795

No gene




associated


796

No gene


797
no gene associated
No gene




associated


798

No gene




associated


799
no gene
No gene




associated


800

No gene




associated


801

No gene




associated


802

No gene




associated


803

No gene




associated


804
sorting nexin 5
SNX5
FLJ10931
NM_014426
OTTHUMG00000031953
27131


805
Probable D-tyrosyl-
HARS2
DUEB;
NM_080820
ENSG00000125821
92675



tRNA(Tyr) deacylase

C20orf88;



(EC 3.1.—.—)

MGC41905;





MGC119131;





bA379J5.3;





bA555E18.1


806
solute carrier family
SLC24A3
NCKX3
NM_020689
OTTHUMG00000031993
57419



24



(sodiumVpotassiumVcalcium



exchanger),



member 3,



OTTHUMG00000031993,



SLC24A3


807

CT026_HUMAN
C20orf26,
NM_015585.2
ENSG00000089101
26074





dJ1178H5.4;





DKFZP434K156


808
RNA-binding protein
RALY
P542;
NM_007367
ENSG00000125970
22913



Raly (hnRNP

MGC117312



associated with lethal



yellow homolog), D



ENSG00000125970,



RALY


809
Protein phosphatase 1
PPP1R16B
TIMAP;
NM_015568
ENSG00000101445
26051



regulatory inhibitor

ANKRD4;



subunit 16B (TGF-

KIAA0823



beta-inhibited



membrane-associated



protein) (hTIMAP)



(CAAX box protein



TIMAP) (Ankyrin



repeat domain protein



4)


810
protein tyrosine
PTPRT
RPTPrho;
NM_007050
OTTHUMG00000033040
11122



phosphatase, receptor

KIAA0283



type, T


811
protein tyrosine
PTPRT
RPTPrho;
NM_007050
OTTHUMG00000033040
11122



phosphatase, receptor

KIAA0283



type, T


812
protein tyrosine
PTPRT
RPTPrho;
NM_007050
OTTHUMG00000033040
11122



phosphatase, receptor

KIAA0283



type, T


813
Receptor-type
PTPRT
RPTPrho;
NM_007050
ENSG00000196090
11122



tyrosine-protein

KIAA0283



phosphatase T



precursor (EC



3.1.3.48) (R-PTP-T)



(RPTP-rho)


814
cadherin-like 22
CDH22
C20orf25;
NM_021248
OTTHUMG00000033073
64405





MGC39564;





dJ998H6.1


815
potassium voltage-
KCNB1
DRK1;
NM_004975
OTTHUMG00000033051
3745



gated channel, Shab-

KV2.1; h-



related subfamily,

DRK1



member 1


816
potassium voltage-
KCNB1
DRK1;
NM_004975
OTTHUMG00000033051
3745



gated channel, Shab-

KV2.1; h-



related subfamily,

DRK1



member 1


817
Zinc finger protein
SNAI1
SNA;
NM_005985
ENSG00000124216
6615



SNAI1 (Snail protein

SNAH;



homolog) (Sna

SLUGH2;



protein)

dJ710H13.1


818
Cadherin-4 precursor
CDH4
CAD4;
NM_001794
ENSG00000179242
1002



(Retinal-cadherin) (R-

RCAD;



cadherin) (R-CAD)

FLJ22202;





FLJ40547;





MGC126700


819
cadherin 4, type 1, R-
CDH4
CAD4;
NM_001794
OTTHUMG00000032890
1002



cadherin (retinal)

RCAD;





FLJ22202;





FLJ40547;





MGC126700


820
Cadherin-4 precursor
CDH4
CAD4;
NM_001794
ENSG00000179242
1002



(Retinal-cadherin) (R-

RCAD;



cadherin) (R-CAD)

FLJ22202;





FLJ40547;





MGC126700


821
Metalloproteinase



inhibitor 3 precursor



(TIMP-3) (Tissue



inhibitor of



metalloproteinases-3)



(MIG-5 protein).


822
Tubulin alpha-8 chain
TUBA8
TUBAL2
NM_018943
ENSG00000070490
51807



(Alpha-tubulin 8)


823

No gene




associated


824

No gene




associated
















TABLE 6





Overview of the genomic regions and ranges of methylation within which the actual value of


methylation of said genomic regions lies specific of the said cells, tissues and/or organs.


The actual value of methylation is shown in FIGS. 1.1-1.403.





















SEQ








ID
CD4
CD8

Embryonic


NO:
T-
T-
Embryonic
Skeletal

Heart


Genomic
lymphocyte
lymphocyte
Liver
Muscle
Fibroblast
Muscle





413
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


414
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


415
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


416
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


417
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


418
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


419
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


420
75-100%
75-100%
75-100%
 0-25%
75-100%
75-100%


421
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


422
25-75%
25-75%
25-75%
25-75%
 0-25%
25-75%


423
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


424
75-100%
75-100%
75-100%
25-75%
75-100%
25-75%


425
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


426
25-75%
25-75%
 0-25%
 0-25%
 0-25%
 0-25%


427
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


428
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


429
 0-25%
 0-25%
 0-25%
75-100%
75-100%
25-75%


430
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


431
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


432
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


433
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


434
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


435
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


436
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


437
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


438
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


439
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


440
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


441
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


442
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


443
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


444
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


445
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


446
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


447
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


448
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


449
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


450
25-75%
25-75%
25-75%
25-75%
75-100%
75-100%


451
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


452
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


453
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


454
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


455
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


456
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


457
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


458
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


459
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%


460
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


461
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


462
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


463
 0-25%
 0-25%
25-75%
25-75%
25-75%
25-75%


464
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


465
75-100%
75-100%
75-100%
 0-25%
 0-25%
 0-25%


466
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


467
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


468
75-100%
75-100%
25-75%
25-75%
 0-25%
25-75%


469
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


470
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


471
75-100%
75-100%
75-100%
75-100%
25-75%
25-75%


472
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


473
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


474
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


475
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%


476
75-100%
75-100%
25-75%
25-75%
75-100%
75-100%


477
75-100%
75-100%
25-75%
 0-25%
 0-25%
25-75%


478
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


479
75-100%
75-100%
25-75%
 0-25%
 0-25%
25-75%


480
25-75%
25-75%
25-75%
75-100%
75-100%
75-100%


481
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


482
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


483
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


484
75-100%
75-100%
25-75%
 0-25%
 0-25%
25-75%


485
75-100%
75-100%
75-100%
 0-25%
 0-25%
 0-25%


486
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


487
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%


488
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


489
 0-25%
 0-25%
25-75%
25-75%
25-75%
25-75%


490
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


491
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


492
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


493
 0-25%
 0-25%
25-75%
25-75%
75-100%
75-100%


494
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


495
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


496
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


497
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


498
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


499
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


500
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


501
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


502
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


503
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


504
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


505
 0-25%
 0-25%
75-100%
 0-25%
 0-25%
 0-25%


506
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


507
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


508
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


509
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%


510
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


511
25-75%
75-100%
75-100%
75-100%
75-100%
25-75%


512
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


513
25-75%
25-75%
75-100%
75-100%
75-100%
75-100%


514
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


515
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


516
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


517
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


518
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


519
 0-25%
 0-25%
25-75%
25-75%
25-75%
25-75%


520
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


521
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


522
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


523
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


524
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


525
 0-25%
 0-25%
ND
 0-25%
75-100%
 0-25%


526
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


527
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


528
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


529
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


530
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


531
 0-25%
 0-25%
25-75%
25-75%
25-75%
25-75%


532
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


533
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


534
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


535
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


536
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


537
25-75%
25-75%
 0-25%
 0-25%
 0-25%
25-75%


538
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


539
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


540
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


541
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


542
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


543
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


544
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


545
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


546
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


547
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


548
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


549
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


550
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


551
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


552
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%


553
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


554
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


555
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


556
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


557
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


558
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


559
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


560
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


561
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


562
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


563
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


564
25-75%
25-75%
25-75%
25-75%
75-100%
25-75%


565
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


566
75-100%
75-100%
ND
25-75%
 0-25%
25-75%


567
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


568
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


569
75-100%
75-100%
75-100%
75-100%
 0-25%
25-75%


570
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


571
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


572
25-75%
25-75%
25-75%
25-75%
75-100%
25-75%


573
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


574
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


575
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


576
 0-25%
 0-25%
 0-25%
 0-25%
75-100%
 0-25%


577
75-100%
75-100%
ND
75-100%
75-100%
75-100%


578
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


579
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
25-75%


580
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


581
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


582
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


583
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


584
 0-25%
 0-25%
 0-25%
 0-25%
75-100%
 0-25%


585
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


586
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


587
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


588
75-100%
75-100%
25-75%
 0-25%
 0-25%
25-75%


589
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


590
25-75%
25-75%
 0-25%
 0-25%
 0-25%
25-75%


591
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


592
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


593
75-100%
75-100%
25-75%
 0-25%
 0-25%
25-75%


594
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


595
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


596
75-100%
75-100%
75-100%
25-75%
 0-25%
75-100%


597
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


598
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


599
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


600
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


601
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


602
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


603
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%


604
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


605
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


606
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


607
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


608
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


609
75-100%
ND
ND
ND
 0-25%
75-100%


610
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


611
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


612
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


613
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


614
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


615
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


616
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


617
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


618
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


619
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


620
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


621
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


622
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


623
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


624
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


625
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


626
75-100%
75-100%
25-75%
25-75%
 0-25%
75-100%


627
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


628
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


629
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


630
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


631
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


632
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


633
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


634
75-100%
25-75%
25-75%
25-75%
25-75%
25-75%


635
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


636
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


637
25-75%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


638
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


639
25-75%
25-75%
75%-100%
75-100%
 0-25%
75-100%


640
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


641
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


642
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


643
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


644
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


645
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


646
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


647
75-100%
75-100%
75-100%
25-75%
 0-25%
75-100%


648
25-75%
25-75%
75-100%
75-100%
 0-25%
75-100%


649
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


650
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


651
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


652
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%


653
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


654
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


655
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


656
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


657
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


658
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


659
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


660
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


661
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


662
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


663
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


664
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


665
75-100%
75-100%
75-100%
ND
25-75%
25-75%


666
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


667
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


668
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


669
75-100%
75-100%
25-75%
25-75%
 0-25%
 0-25%


670
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


671
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


672
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


673
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


674
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


675
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


676
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


677
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


678
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


679
 0-25%
 0-25%
 0-25%
75-100%
75-100%
75-100%


680
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


681
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


682
75-100%
75-100%
 0-25%
 0-25%
 0-25%
25-75%


683
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


684
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


685
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


686
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


687
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


688
25-75%
25-75%
25-75%
25-75%
 0-25%
25-75%


689
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


690
75-100%
75-100%
75-100%
25-75%
 0-25%
75-100%


691
75-100%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


692
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


693
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


694
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


695
 0-25%
 0-25%
25-75%
75-100%
75-100%
75-100%


696
25-75%
25-75%
25-75%
25-75%
25-75%
 0-25%


697
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


698
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


699
75-100%
75-100%
ND
ND
25-75%
75-100%


700
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


701
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


702
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


703
25-75%
25-75%
75-100%
75-100%
75-100%
75-100%


704
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


705
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


706
25-75%
75-100%
25-75%
25-75%
25-75%
25-75%


707
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


708
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


709
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


710
 0-25%
 0-25%
25-75%
25-75%
75-100%
75-100%


711
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


712
75-100%
25-75%
25-75%
75-100%
75-100%
75-100%





SEQ


ID


NO:




Skeletal


Genomic
Keratinocyte
Liver
Melanocyte
Placenta
Muscle
Sperm





413
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


414
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


415
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


416
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


417
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


418
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%


419
75-100%
75-100%
75-100%
 0-25%
75-100%
 0-25%


420
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


421
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


422
75-100%
25-75%
75-100%
25-75%
25-75%
 0-25%


423
 0-25%
 0-25%
75-100%
 0-25%
 0-25%
75-100%


424
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%


425
25-75%
75-100%
75-100%
25-75%
25-75%
75-100%


426
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


427
75-100%
25-75%
75-100%
75-100%
75-100%
 0-25%


428
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


429
25-75%
 0-25%
 0-25%
75-100%
75-100%
 0-25%


430
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


431
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


432
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


433
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%


434
75-100%
25-75%
75-100%
75-100%
75-100%
 0-25%


435
25-75%
75-100%
75-100%
75-100%
75-100%
 0-25%


436
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


437
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


438
25-75%
25-75%
25-75%
25-75%
25-75%
 0-25%


439
75-100%
25-75%
25-75%
25-75%
25-75%
25-75%


440
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


441
75-100%
75-100%
ND
75-100%
25-75%
75-100%


442
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


443
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%


444
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


445
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


446
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


447
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


448
25-75%
75-100%
25-75%
25-75%
25-75%
75-100%


449
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


450
 0-25%
25-75%
75-100%
25-75%
25-75%
ND


451
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


452
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


453
75-100%
75-100%
75-100%
75-100%
 0-25%
 0-25%


454
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%


455
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%


456
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


457
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


458
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


459
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


460
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


461
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


462
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%


463
 0-25%
25-75%
25-75%
25-75%
25-75%
75-100%


464
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


465
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


466
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


467
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


468
 0-25%
25-75%
25-75%
25-75%
25-75%
75-100%


469
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


470
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


471
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%


472
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


473
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


474
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


475
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


476
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


477
 0-25%
25-75%
 0-25%
25-75%
25-75%
75-100%


478
25-75%
25-75%
 0-25%
25-75%
25-75%
75-100%


479
 0-25%
25-75%
 0-25%
 0-25%
75-100%
75-100%


480
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


481
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


482
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


483
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


484
 0-25%
25-75%
 0-25%
 0-25%
 0-25%
75-100%


485
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


486
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


487
75-100%
75-100%
 0-25%
25-75%
75-100%
75-100%


488
75-100%
75-100%
 0-25%
25-75%
25-75%
75-100%


489
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


490
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


491
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


492
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


493
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


494
 0-25%
75-100%
75-100%
75-100%
75-100%
 0-25%


495
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


496
25-75%
25-75%
 0-25%
25-75%
25-75%
 0-25%


497
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


498
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


499
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


500
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


501
75-100%
25-75%
25-75%
25-75%
25-75%
 0-25%


502
25-75%
25-75%
25-75%
25-75%
75-100%
 0-25%


503
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


504
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


505
 0-25%
75-100%
 0-25%
 0-25%
 0-25%
ND


506
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


507
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


508
 0-25%
75-100%
 0-25%
 0-25%
 0-25%
ND


509
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


510
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


511
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


512
 0-25%
75-100%
75-100%
25-75%
25-75%
75-100%


513
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


514
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


515
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


516
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


517
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


518
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


519
25-75%
 0-25%
25-75%
25-75%
75-100%
 0-25%


520
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


521
 0-25%
75-100%
75-100%
75-100%
75-100%
 0-25%


522
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


523
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%


524
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


525
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


526
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


527
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


528
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


529
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


530
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


531
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


532
 0-25%
75-100%
75-100%
75-100%
75-100%
 0-25%


533
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


534
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


535
75-100%
75-100%
ND
75-100%
75-100%
75-100%


536
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


537
 0-25%
75-100%
 0-25%
 0-25%
 0-25%
75-100%


538
 0-25%
 0-25%
 0-25%
25-75%
 0-25%
 0-25%


539
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


540
75-100%
 0-25%
75-100%
75-100%
75-100%
75-100%


541
 0-25%
25-75%
25-75%
25-75%
25-75%
 0-25%


542
 0-25%
25-75%
25-75%
25-75%
25-75%
 0-25%


543
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


544
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


545
 0-25%
 0-25%
25-75%
ND
 0-25%
10-25%


546
 0-25%
75-100%
25-75%
 0-25%
 0-25%
75-100%


547
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


548
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


549
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%


550
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


551
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


552
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


553
75-100%
25-75%
75-100%
75-100%
75-100%


554
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


555
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


556
 0-25%
25-75%
75-100%
75-100%
75-100%
75-100%


557
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


558
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


559
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


560
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


561
 0-25%
25-75%
25-75%
25-75%
25-75%
75-100%


562
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


563
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


564
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%


565
 0-25%
75-100%
75-100%
75-100%
75-100%
 0-25%


566
 0-25%
25-75%
25-75%
25-75%
25-75%
ND


567
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


568
 0-25%
25-75%
 0-25%
 0-25%
 0-25%
 0-25%


569
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


570
 0-25%
75-100%
75-100%
75-100%
75-100%
 0-25%


571
 0-25%
25-75%
25-75%
25-75%
25-75%
75-100%


572
75-100%
25-75%
75-100%
25-75%
25-75%
75-100%


573
75-100%
75-100%
75-100%
75-100%
25-75%
 0-25%


574
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


575
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


576
75-100%
 0-25%
75-100%
 0-25%
 0-25%
 0-25%


577
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


578
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


579
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


580
75-100%
75-100%
75-100%
75-100%
25-75%
 0-25%


581
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


582
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


583
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


584
 0-25%
 0-25%
 0-25%
 0-25%
25-75%
75-100%


585
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


586
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


587
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


588
 0-25%
25-75%
 0-25%
 0-25%
 0-25%
 0-25%


589
75-100%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


590
 0-25%
75-100%
75-100%
 0-25%
 0-25%
 0-25%


591
 0-25%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%


592
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%


593
25-75%
25-75%
25-75%
 0-25%
 0-25%
 0-25%


594
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


595
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


596
75-100%
75-100%
 0-25%
25-75%
75-100%
75-100%


597
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


598
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


599
75-100%
25-75%
75-100%
75-100%
25-75%
75-100%


600
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%


601
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


602
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


603
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


604
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%


605
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%


606
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


607
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


608
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


609
75-100%
75-100%
 0-25%
 0-25%
75-100%
75-100%


610
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


611
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


612
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


613
75-100%
75-100%
 0-25%
 0-25%
 0-25%
ND


614
75-100%
25-75%
25-75%
25-75%
25-75%
75-100%


615
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


616
25-75%
75-100%
25-75%
25-75%
25-75%
75-100%


617
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


618
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%


619
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


620
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


621
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


622
25-75%
75-100%
75-100%
75-100%
 0-25%
75-100%


623
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


624
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


625
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


626
75-100%
75-100%
75-100%
25-75%
25-75%
 0-25%


627
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%


628
 0-25%
75-100%
75-100%
25-75%
25-75%
75-100%


629
25-75%
25-75%
25-75%
ND
75-100%
75-100%


630
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


631
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


632
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


633
75-100%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


634
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


635
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


636
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


637
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


638
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%


639
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


640
25-75%
75-100%
25-75%
75-100%
75-100%
75-100%


641
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


642
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


643
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


644
75-100%
75-100%
 0-25%
25-75%
25-75%
75-100%


645
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


646
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


647
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


648
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


649
 0-25%
75-100%
75-100%
75-100%
75-100%


650
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%


651
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


652
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


653
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


654
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


655
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


656
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%


657
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


658
75-100%
75-100%
75-100%
ND
25-75%
ND


659
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


660
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


661
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


662
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


663
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


664
75-100%
75-100%
75-100%
75-100%
 0-25%
ND


665
25-75%
25-75%
25-75%
25-75%
25-75%
 0-25%


666
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


667
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


668
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


669
 0-25%
25-75%
 0-25%
 0-25%
25-75%
 0-25%


670
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


671
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%


672
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%


673
25-75%
75-100%
25-75%
25-75%
 0-25%
ND


674
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%


675
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


676
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
75-100%


677
75-100%
 0-25%
 0-25%
 0-25%
 0-25%
ND


678
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


679
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


680
25-75%
75-100%
25-75%
25-75%
25-75%
25-75%


681
 0-25%
 0-25%
75-100%
 0-25%
 0-25%
75-100%


682
 0-25%
25-75%
 0-25%
 0-25%
25-75%
75-100%


683
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


684
 0-25%
25-75%
 0-25%
 0-25%
 0-25%
 0-25%


685
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


686
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


687
75-100%
25-75%
25-75%
25-75%
25-75%
75-100%


688
25-75%
25-75%
25-75%
25-75%
25-75%
ND


689
 0-25%
75-100%
75-100%
75-100%
75-100%
 0-25%


690
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


691
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


692
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


693
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


694
75-100%
75-100%
75-100%
75-100%
75-100%
 0-25%


695
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


696
75-100%
25-75%
25-75%
25-75%
 0-25%
 0-25%


697
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


698
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


699
75-100%
75-100%
75-100%
ND
75-100%
75-100%


700
 0-25%
25-75%
 0-25%
 0-25%
25-75%
75-100%


701
75-100%
 0-25%
75-100%
75-100%
75-100%
75-100%


702
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


703
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


704
25-75%
25-75%
25-75%
25-75%
75-100%
75-100%


705
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


706
25-75%
75-100%
25-75%
25-75%
25-75%
75-100%


707
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


708
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


709
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%


710
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


711
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%


712
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%





ND = not determined













TABLE 7





Overview of the genomic regions and ranges of methylation within which the actual value of


methylation of said genomic regions lies specific of the said cells, tissues and/or organs.


The actual value of methylation is shown in FIGS. 1.1-1.403.





















SEQ








ID
CD4
CD8

Embryonic


NO:
T-
T-
Embryonic
Skeletal

Heart


Genomic
lymphocyte
lymphocyte
Liver
Muscle
Fibroblast
Muscle





713
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


714
25-75%
25-75%
ND
 0-25%
 0-25%
25-75%


715
25-75%
25-75%
 0-25%
 0-25%
 0-25%
 0-25%


716
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


717
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


718
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


719
25-75%
25-75%
25-75%
 0-25%
 0-25%
25-75%


720
 0-25%
 0-25%
 0-25%
75-100%
75-100%
 0-25%


721
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


722
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


723
75-100%
75-100%
75-100%
 0-25%
 0-25%
75-100%


724
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


725
75-100%
75-100%
75-100%
75-100%
25-75%
25-75%


726
 0-25%
 0-25%
25-75%
 0-25%
 0-25%
25-75%


727
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


728
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


729
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


730
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%


731
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


732
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


733
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


734
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


735
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


736
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


737
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


738
 0-25%
 0-25%
 0-25%
 0-25%
25-75%
25-75%


739
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


740
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


741
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


742
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


743
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


744
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


745
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


746
75-100%
75-100%
75-100%
 0-25%
 0-25%
25-75%


747
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


748
75-100%
75-100%
75-100%
25-75%
75-100%
75-100%


749
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


750
75-100%
75-100%
25-75%
25-75%
 0-25%
 0-25%


751
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


752
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


753
75-100%
75-100%
75-100%
ND
 0-25%
75-100%


754
75-100%
75-100%
25-75%
25-75%
25-75%
75-100%


755
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


756
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


757
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


758
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


759
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


760
 0-25%
 0-25%
25-75%
25-75%
25-75%
25-75%


761
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


762
 0-25%
ND
 0-25%
75-100%
75-100%
75-100%


763
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%


764
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


765
 0-25%
 0-25%
 0-25%
25-75%
25-75%
25-75%


766
 0-25%
 0-25%
25-75%
75-100%
75-100%
75-100%


767
25-75%
25-75%
25-75%
 0-25%
 0-25%
 0-25%


768
25-75%
25-75%
 0-25%
25-75%
25-75%
25-75%


769
75-100%
75-100%
75-100%
25-75%
 0-25%
25-75%


770
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


771
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


772
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%


773
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


774
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


775
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


776
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


777
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


778
 0-25%
 0-25%
75-100%
75-100%
75-100%
75-100%


779
75-100%
75-100%
75-100%
ND
25-75%
25-75%


780
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


781
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%


782
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


783
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


784
75-100%
75-100%
75-100%
75-100%
25-75%
25-75%


785
25-75%
25-75%
25-75%
25-75%
 0-25%
 0-25%


786
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


787
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%
 0-25%


788
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


789
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


790
25-75%
25-75%
25-75%
25-75%
25-75%
25-75%


791
 0-25%
 0-25%
 0-25%
 0-25%
75-100%
25-75%


792
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%


793
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


794
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


795
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%


796
75-100%
75-100%
75-100%
 0-25%
 0-25%
25-75%


797
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


798
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


799
75-100%
75-100%
75-100%
 0-25%
 0-25%
 0-25%


800
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


801
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


802
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


803
75-100%
75-100%
75-100%
ND
 0-25%
25-75%


804
25-75%
25-75%
25-75%
25-75%
25-75%
75-100%


805
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


806
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


807
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


808
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


809
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


810
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


811
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


812
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%


813
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


814
75-100%
75-100%
75-100%
75-100%

75-100%


815
75-100%
75-100%
75-100%
25-75%
25-75%
25-75%


816
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%


817
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%


818
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%


819
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%


820
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


821
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


822
25-75%
25-75%
25-75%
25-75%
75-100%
25-75%


823
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%


824
75-100%
75-100%
75-100%
75-100%
75-100%
25-75%


















SEQ









ID



NO:




Skeletal



Genomic
Keratinocyte
Liver
Melanocyte
Placenta
Muscle
Sperm







713
75-100%
25-75%
75-100%
75-100%
25-75%
25-75%



714
 0-25%
25-75%
 0-25%
25-75%
25-75%
25-75%



715
 0-25%
25-75%
 0-25%
 0-25%
 0-25%
 0-25%



716
25-75%
25-75%
75-100%
25-75%
25-75%
 0-25%



717
25-75%
75-100%
 0-25%
25-75%
25-75%
 0-25%



718
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%



719
 0-25%
25-75%
 0-25%
25-75%
25-75%
 0-25%



720
 0-25%
 0-25%
 0-25%
75-100%
75-100%
 0-25%



721
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%



722
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%



723
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%



724
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%



725
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



726
 0-25%
25-75%
25-75%
25-75%
 0-25%
75-100%



727
75-100%
25-75%
75-100%
25-75%
25-75%
25-75%



728
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%



729
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%



730
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



731
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%



732
75-100%
75-100%
25-75%
75-100%
25-75%
75-100%



733
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



734
 0-25%
 0-25%
75-100%
 0-25%
 0-25%
 0-25%



735
25-75%
75-100%
75-100%
25-75%
25-75%
75-100%



736
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%



737
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



738
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



739
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%



740
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%



741
 0-25%
75-100%
 0-25%
25-75%
25-75%
75-100%



742
25-75%
75-100%
75-100%
25-75%
25-75%
75-100%



743
 0-25%
75-100%
75-100%
 0-25%
75-100%
 0-25%



744
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%



745
 0-25%
75-100%
75-100%
75-100%
 0-25%
 0-25%



746
 0-25%
75-100%
 0-25%
 0-25%
25-75%
75-100%



747
25-75%
75-100%
75-100%
75-100%
25-75%
75-100%



748
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%



749
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%



750
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%



751
 0-25%
75-100%
 0-25%
75-100%
 0-25%
 0-25%



752
75-100%
25-75%
75-100%
75-100%
25-75%
75-100%



753
75-100%
75-100%
75-100%
 0-25%
 0-25%
75-100%



754
25-75%
75-100%
 0-25%
 0-25%
 0-25%
 0-25%



755
75-100%
75-100%
75-100%
25-75%
25-75%
 0-25%



756
 0-25%
75-100%
75-100%
75-100%
25-75%
75-100%



757
25-75%
75-100%
25-75%
25-75%
25-75%
75-100%



758
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%



759
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



760
25-75%
75-100%
25-75%
25-75%
25-75%
75-100%



761
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%



762
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%



763
75-100%
25-75%
75-100%
75-100%
25-75%
75-100%



764
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



765
 0-25%
25-75%
25-75%
25-75%
75-100%
 0-25%



766
75-100%
25-75%
75-100%
25-75%
75-100%
75-100%



767
25-75%
25-75%
25-75%
 0-25%
 0-25%
75-100%



768
 0-25%
25-75%
 0-25%
25-75%
25-75%
 0-25%



769
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



770
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%



771
25-75%
75-100%
75-100%
75-100%
25-75%
75-100%



772
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



773
 0-25%
75-100%
 0-25%
75-100%
75-100%
75-100%



774
75-100%
25-75%
75-100%
75-100%
75-100%
75-100%



775
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%



776
25-75%
75-100%
75-100%
75-100%
75-100%
75-100%



777
 0-25%
75-100%
 0-25%
75-100%
75-100%
75-100%



778
 0-25%
75-100%
75-100%
75-100%
75-100%
75-100%



779
75-100%
75-100%
 0-25%
75-100%
25-75%
75-100%



780
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



781
25-75%
75-100%
25-75%
25-75%
75-100%
75-100%



782
25-75%
75-100%
25-75%
75-100%
75-100%
75-100%



783
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



784
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



785
25-75%
25-75%
 0-25%
 0-25%
 0-25%
75-100%



786
25-75%
25-75%
25-75%
25-75%
25-75%
 0-25%



787
 0-25%
75-100%
 0-25%
 0-25%
75-100%
75-100%



788
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



789
75-100%
75-100%
 0-25%
75-100%
75-100%
75-100%



790
25-75%
75-100%
25-75%
25-75%
 0-25%
75-100%



791
 0-25%
75-100%
25-75%
75-100%
25-75%
75-100%



792
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



793
75-100%
25-75%
25-75%
75-100%
75-100%
75-100%



794
75-100%
25-75%
25-75%
75-100%
75-100%
75-100%



795
75-100%
25-75%
25-75%
25-75%
25-75%
ND



796
75-100%
25-75%
 0-25%
25-75%
25-75%
 0-25%



797
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



798
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



799
 0-25%
75-100%
 0-25%
 0-25%
 0-25%
75-100%



800
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



801
75-100%
25-75%
75-100%
25-75%
25-75%
75-100%



802
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



803
25-75%
75-100%
 0-25%
75-100%
75-100%
75-100%



804
75-100%
75-100%
25-75%
25-75%
25-75%
25-75%



805
75-100%
75-100%
25-75%
25-75%
25-75%
75-100%



806
75-100%
75-100%
25-75%
25-75%
25-75%
75-100%



807
 0-25%
75-100%
75-100%
 0-25%
 0-25%
75-100%



808
25-75%
75-100%
75-100%
25-75%
25-75%
75-100%



809
75-100%
75-100%
75-100%
75-100%
75-100%
75-100%



810
75-100%
25-75%
25-75%
75-100%
75-100%
75-100%



811
75-100%
75-100%
25-75%
25-75%
25-75%
75-100%



812
75-100%
75-100%
25-75%
25-75%
25-75%
75-100%



813
75-100%
75-100%
25-75%
75-100%
75-100%
75-100%



814
75-100%
75-100%
75-100%

25-75%
75-100%



815
75-100%
75-100%
25-75%
25-75%
25-75%
75-100%



816
25-75%
75-100%
75-100%
25-75%
25-75%
75-100%



817
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



818
75-100%
75-100%
75-100%
75-100%
 0-25%
75-100%



819
75-100%
75-100%
25-75%
25-75%
25-75%
75-100%



820
 0-25%
75-100%
75-100%
 0-25%
 0-25%
75-100%



821
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



822
75-100%
75-100%
75-100%
75-100%
25-75%
75-100%



823
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%



824
75-100%
75-100%
75-100%
25-75%
25-75%
75-100%







ND = not determined













TABLE 8A







Sequences for the differentiation and/or detection of T-lymphocytes.








SEQ ID NO:



Genomic
Tissue





511
CD4 T-lymphocyte


634
CD4 T-lymphocyte


637
CD4 T-lymphocyte


652
CD4 T-lymphocyte


416
CD4 T-lymphocyte, CD8 T-



lymphocyte


417
CD4 T-lymphocyte, CD8 T-



lymphocyte


426
CD4 T-lymphocyte, CD8 T-



lymphocyte


438
CD4 T-lymphocyte, CD8 T-



lymphocyte


442
CD4 T-lymphocyte, CD8 T-



lymphocyte


445
CD4 T-lymphocyte, CD8 T-



lymphocyte


449
CD4 T-lymphocyte, CD8 T-



lymphocyte


451
CD4 T-lymphocyte, CD8 T-



lymphocyte


452
CD4 T-lymphocyte, CD8 T-



lymphocyte


458
CD4 T-lymphocyte, CD8 T-



lymphocyte


467
CD4 T-lymphocyte, CD8 T-



lymphocyte


468
CD4 T-lymphocyte, CD8 T-



lymphocyte


474
CD4 T-lymphocyte, CD8 T-



lymphocyte


481
CD4 T-lymphocyte, CD8 T-



lymphocyte


482
CD4 T-lymphocyte, CD8 T-



lymphocyte


483
CD4 T-lymphocyte, CD8 T-



lymphocyte


489
CD4 T-lymphocyte, CD8 T-



lymphocyte


490
CD4 T-lymphocyte, CD8 T-



lymphocyte


491
CD4 T-lymphocyte, CD8 T-



lymphocyte


492
CD4 T-lymphocyte, CD8 T-



lymphocyte


493
CD4 T-lymphocyte, CD8 T-



lymphocyte


497
CD4 T-lymphocyte, CD8 T-



lymphocyte


513
CD4 T-lymphocyte, CD8 T-



lymphocyte


515
CD4 T-lymphocyte, CD8 T-



lymphocyte


518
CD4 T-lymphocyte, CD8 T-



lymphocyte


522
CD4 T-lymphocyte, CD8 T-



lymphocyte


527
CD4 T-lymphocyte, CD8 T-



lymphocyte


528
CD4 T-lymphocyte, CD8 T-



lymphocyte


531
CD4 T-lymphocyte, CD8 T-



lymphocyte


535
CD4 T-lymphocyte, CD8 T-



lymphocyte


542
CD4 T-lymphocyte, CD8 T-



lymphocyte


554
CD4 T-lymphocyte, CD8 T-



lymphocyte


555
CD4 T-lymphocyte, CD8 T-



lymphocyte


557
CD4 T-lymphocyte, CD8 T-



lymphocyte


558
CD4 T-lymphocyte, CD8 T-



lymphocyte


562
CD4 T-lymphocyte, CD8 T-



lymphocyte


566
CD4 T-lymphocyte, CD8 T-



lymphocyte


574
CD4 T-lymphocyte, CD8 T-



lymphocyte


588
CD4 T-lymphocyte, CD8 T-



lymphocyte


597
CD4 T-lymphocyte, CD8 T-



lymphocyte


598
CD4 T-lymphocyte, CD8 T-



lymphocyte


607
CD4 T-lymphocyte, CD8 T-



lymphocyte


632
CD4 T-lymphocyte, CD8 T-



lymphocyte


639
CD4 T-lymphocyte, CD8 T-



lymphocyte


651
CD4 T-lymphocyte, CD8 T-



lymphocyte


659
CD4 T-lymphocyte, CD8 T-



lymphocyte


669
CD4 T-lymphocyte, CD8 T-



lymphocyte


675
CD4 T-lymphocyte, CD8 T-



lymphocyte


676
CD4 T-lymphocyte, CD8 T-



lymphocyte


682
CD4 T-lymphocyte, CD8 T-



lymphocyte


683
CD4 T-lymphocyte, CD8 T-



lymphocyte


685
CD4 T-lymphocyte, CD8 T-



lymphocyte


691
CD4 T-lymphocyte, CD8 T-



lymphocyte


695
CD4 T-lymphocyte, CD8 T-



lymphocyte


697
CD4 T-lymphocyte, CD8 T-



lymphocyte


710
CD4 T-lymphocyte, CD8 T-



lymphocyte


463
CD4 T-lymphocyte, CD8 T-



lymphocyte


465
CD4 T-lymphocyte, CD8 T-



lymphocyte


679
CD4 T-lymphocyte, CD8 T-



lymphocyte


480
CD4 T-lymphocyte, CD8 T-



lymphocyte


701
CD4 T-lymphocyte, CD8 T-



lymphocyte


706
CD8 T-lymphocyte
















TABLE 8B







Sequences for the differentiation and/or detection of embryonic liver.








SEQ ID NO:



Genomic
Tissue





509
embryonic



liver


459
embryonic



liver


475
embryonic



liver


476
embryonic



liver


477
embryonic



liver


479
embryonic



liver


484
embryonic



liver


485
embryonic



liver


552
embryonic



liver


593
embryonic



liver


596
embryonic



liver


654
embryonic



liver


712
embryonic



liver


429
embryonic



liver


465
embryonic



liver


679
embryonic



liver


480
embryonic



liver


420
embryonic



liver


612
embryonic



liver


621
embryonic



liver


624
embryonic



liver


678
embryonic



liver


505
embryonic



liver
















TABLE 8C







Sequences for the differentiation and/or detection of embryonic skeletal


muscle.








SEQ ID NO:



Genomic
Tissue





444
embryonic skeletal



muscle


658
embryonic skeletal



muscle


441
embryonic skeletal



muscle


459
embryonic skeletal



muscle


475
embryonic skeletal



muscle


476
embryonic skeletal



muscle


477
embryonic skeletal



muscle


479
embryonic skeletal



muscle


484
embryonic skeletal



muscle


485
embryonic skeletal



muscle


552
embryonic skeletal



muscle


593
embryonic skeletal



muscle


596
embryonic skeletal



muscle


654
embryonic skeletal



muscle


712
embryonic skeletal



muscle


429
embryonic skeletal



muscle


465
embryonic skeletal



muscle


679
embryonic skeletal



muscle


480
embryonic skeletal



muscle


420
embryonic skeletal



muscle


612
embryonic skeletal



muscle


621
embryonic skeletal



muscle


624
embryonic skeletal



muscle


678
embryonic skeletal



muscle
















TABLE 8D







Sequences for the differentiation and/or detection of fibroblasts:








SEQ ID NO:



Genomic
Tissue





422
fibroblast


440
fibroblast


446
fibroblast


473
fibroblast


504
fibroblast


524
fibroblast


534
fibroblast


563
fibroblast


567
fibroblast


569
fibroblast


580
fibroblast


584
fibroblast


586
fibroblast


594
fibroblast


601
fibroblast


610
fibroblast


611
fibroblast


617
fibroblast


626
fibroblast


630
fibroblast


631
fibroblast


636
fibroblast


638
fibroblast


641
fibroblast


642
fibroblast


643
fibroblast


645
fibroblast


646
fibroblast


647
fibroblast


648
fibroblast


653
fibroblast


655
fibroblast


660
fibroblast


688
fibroblast


690
fibroblast


699
fibroblast


708
fibroblast


709
fibroblast


471
fibroblast


572
fibroblast


640
fibroblast


576
fibroblast
















TABLE 8E







Sequences for the differentiation and/or detection of heart muscle.








SEQ ID NO:



Genomic
Tissue





430
heart muscle


579
heart muscle


595
heart muscle


615
heart muscle


619
heart muscle


698
heart muscle


700
heart muscle


424
heart muscle
















TABLE 8F







Sequences for the differentiation of heart muscle from skeletal muscle:








SEQ ID NO:



Genomic
Tissue





525
heart muscle versus skeletal muscle


590
heart muscle versus skeletal muscle


599
heart muscle versus skeletal muscle


704
heart muscle versus skeletal muscle
















TABLE 8G







Sequences for the differentiation and/or detection of keratinocytes.








SEQ ID NO:



Genomic
Tissue





463
keratinocyte


433
keratinocyte


435
keratinocyte


439
keratinocyte


447
keratinocyte


450
keratinocyte


454
keratinocyte


460
keratinocyte


461
keratinocyte


462
keratinocyte


494
keratinocyte


495
keratinocyte


499
keratinocyte


501
keratinocyte


506
keratinocyte


512
keratinocyte


516
keratinocyte


520
keratinocyte


521
keratinocyte


526
keratinocyte


532
keratinocyte


533
keratinocyte


536
keratinocyte


539
keratinocyte


541
keratinocyte


549
keratinocyte


550
keratinocyte


551
keratinocyte


556
keratinocyte


559
keratinocyte


561
keratinocyte


564
keratinocyte


565
keratinocyte


570
keratinocyte


571
keratinocyte


575
keratinocyte


587
keratinocyte


589
keratinocyte


592
keratinocyte


600
keratinocyte


602
keratinocyte


603
keratinocyte


604
keratinocyte


605
keratinocyte


608
keratinocyte


609
keratinocyte


614
keratinocyte


625
keratinocyte


627
keratinocyte


628
keratinocyte


633
keratinocyte


635
keratinocyte


649
keratinocyte


650
keratinocyte


662
keratinocyte


663
keratinocyte


666
keratinocyte


667
keratinocyte


670
keratinocyte


677
keratinocyte


687
keratinocyte


689
keratinocyte


692
keratinocyte


696
keratinocyte


702
keratinocyte


703
keratinocyte


707
keratinocyte


709
keratinocyte


471
keratinocyte


572
keratinocyte


640
keratinocyte


576
keratinocyte


613
keratinocyte
















TABLE 8H







Sequences for the differentiation and/or detection of liver.








SEQ



ID NO: Genomic
Tissue





415
liver


427
liver


434
liver


448
liver


456
liver


457
liver


503
liver


508
liver


529
liver


537
liver


540
liver


546
liver


548
liver


553
liver


568
liver


582
liver


583
liver


591
liver


606
liver


616
liver


620
liver


661
liver


680
liver


684
liver


693
liver


705
liver


711
liver


701
liver


613
liver


505
liver
















TABLE 8I







Sequences for the differentiation and/or detection of melanocytes.








SEQ ID NO:



Genomic
Tissue





418
melanocyte


423
melanocyte


443
melanocyte


455
melanocyte


478
melanocyte


487
melanocyte


488
melanocyte


496
melanocyte


523
melanocyte


545
melanocyte


618
melanocyte


644
melanocyte


656
melanocyte


671
melanocyte


672
melanocyte


674
melanocyte


681
melanocyte


471
melanocyte


572
melanocyte


640
melanocyte


576
melanocyte
















TABLE 8J







Sequences for the differentiation and/or detection of placenta.








SEQ ID NO:



Genomic
Tissue





436
placenta


538
placenta


419
placenta
















TABLE 8K







Sequences for the differentiation and/or detection of skeletal muscle.








SEQ ID NO:



Genomic
Tissue





432
skeletal muscle


453
skeletal muscle


466
skeletal muscle


469
skeletal muscle


472
skeletal muscle


498
skeletal muscle


500
skeletal muscle


514
skeletal muscle


517
skeletal muscle


530
skeletal muscle


544
skeletal muscle


547
skeletal muscle


573
skeletal muscle


577
skeletal muscle


585
skeletal muscle


622
skeletal muscle


657
skeletal muscle


664
skeletal muscle


668
skeletal muscle


673
skeletal muscle


686
skeletal muscle


441
skeletal muscle


424
skeletal muscle


502
skeletal muscle


519
skeletal muscle


629
skeletal muscle


420
skeletal muscle


612
skeletal muscle


621
skeletal muscle


624
skeletal muscle


678
skeletal muscle


658
skeletal muscle
















TABLE 8L







Sequences for the differentiation and/or detection of sperm.








SEQ ID NO:



Genomic
Tissue





413
sperm


414
sperm


421
sperm


428
sperm


431
sperm


437
sperm


464
sperm


470
sperm


486
sperm


507
sperm


510
sperm


543
sperm


560
sperm


578
sperm


581
sperm


623
sperm


665
sperm


694
sperm
















TABLE 9A







Sequences for the differentiation and/or detection of T-lymphocytes.








SEQ ID NO:



Genomic
Tissue





730
CD4 T-lymphocyte


760
CD4 T-lymphocyte, CD8 T-



lymphocyte


766
CD4 T-lymphocyte, CD8 T-



lymphocyte


778
CD4 T-lymphocyte, CD8 T-



lymphocyte


786
CD4 T-lymphocyte, CD8 T-



lymphocyte


715
CD4 T-lymphocyte, CD8 T-



lymphocyte


799
CD4 T-lymphocyte, CD8 T-



lymphocyte
















TABLE 9B







Sequences for the differentiation and/or detection of embryonic liver.








SEQ ID NO:



Genomic
Tissue





752
embryonic liver


763
embryonic liver
















TABLE 9C







Sequences for the differentiation and/or detection


of embryonic skeletal muscle.








SEQ ID NO:



Genomic
Tissue





781
embryonic skeletal



muscle


717
embryonic skeletal



muscle


719
embryonic skeletal



muscle


714
embryonic skeletal



muscle
















TABLE 9D







Sequences for the differentiation and/or detection of fibroblasts.








SEQ ID NO:



Genomic
Tissue





720
fibroblast


723
fibroblast


725
fibroblast


733
fibroblast


737
fibroblast


738
fibroblast


753
fibroblast


755
fibroblast


758
fibroblast


761
fibroblast


764
fibroblast


767
fibroblast


768
fibroblast


769
fibroblast


770
fibroblast


774
fibroblast


780
fibroblast


782
fibroblast


784
fibroblast


788
fibroblast


792
fibroblast


801
fibroblast


809
fibroblast


814
fibroblast


817
fibroblast


818
fibroblast


718
fibroblast


741
fibroblast


751
fibroblast


773
fibroblast


823
fibroblast


772
fibroblast


800
fibroblast


821
fibroblast


744
fibroblast


717
fibroblast


719
fibroblast


714
fibroblast
















TABLE 9E







Sequences for the differentiation and/or detection of heart muscle.








SEQ ID NO:



Genomic
Tissue





759
heart muscle


822
heart muscle


824
heart muscle
















TABLE 9F







Sequences for the differentiation and/or detection of keratinocytes.








SEQ ID NO:



Genomic
Tissue





716
keratinocyte


724
keratinocyte


727
keratinocyte


728
keratinocyte


729
keratinocyte


732
keratinocyte


734
keratinocyte


739
keratinocyte


740
keratinocyte


743
keratinocyte


745
keratinocyte


748
keratinocyte


762
keratinocyte


765
keratinocyte


771
keratinocyte


776
keratinocyte


777
keratinocyte


785
keratinocyte


789
keratinocyte


791
keratinocyte


795
keratinocyte


796
keratinocyte


803
keratinocyte


804
keratinocyte


805
keratinocyte


806
keratinocyte


811
keratinocyte


812
keratinocyte


813
keratinocyte


815
keratinocyte


816
keratinocyte


819
keratinocyte


820
keratinocyte


778
keratinocyte


786
keratinocyte


741
keratinocyte


751
keratinocyte


773
keratinocyte


744
keratinocyte


717
keratinocyte


719
keratinocyte


714
keratinocyte


807
keratinocyte
















TABLE 9G







Sequences for the differentiation and/or detection of liver.








SEQ ID NO:



Genomic
Tissue





746
liver


757
liver


793
liver


763
liver


713
liver


787
liver


790
liver


715
liver


799
liver
















TABLE 9H







Sequences for the differentiation and/or detection of melanocytes.








SEQ ID NO:



Genomic
Tissue





721
melanocyte


722
melanocyte


726
melanocyte


731
melanocyte


735
melanocyte


736
melanocyte


742
melanocyte


747
melanocyte


749
melanocyte


750
melanocyte


754
melanocyte


756
melanocyte


775
melanocyte


779
melanocyte


794
melanocyte


808
melanocyte


810
melanocyte


744
melanocyte


717
melanocyte


719
melanocyte


714
melanocyte


718
melanocyte


741
melanocyte


751
melanocyte


773
melanocyte
















TABLE 9I







Sequences for the differentiation and/or detection of placenta.








SEQ ID NO:



Genomic
Tissue





823
placenta


824
placenta


820
placenta
















TABLE 9J







Sequences for the differentiation and/or detection of skeletal muscle.








SEQ ID NO:



Genomic
Tissue





783
skeletal muscle


797
skeletal muscle


798
skeletal muscle


802
skeletal muscle


752
skeletal muscle


823
skeletal muscle


772
skeletal muscle


800
skeletal muscle


821
skeletal muscle


759
skeletal muscle


822
skeletal muscle


824
skeletal muscle


820
skeletal muscle


713
skeletal muscle


787
skeletal muscle


790
skeletal muscle


807
skeletal muscle









EXAMPLES
Example 1

More than 300 specific regions of differential methylation have been identified in primary cells and tissues. Some of them are located in known promoter regions, while others are located in intra- or intergenic regions throughout the chromosomes 20, 22 and 6.


a) Materials and Methods

Samples: Studied primary cell cultures included lymphocytes (selected and sorted by CD4 and CD8 antigen expression), melanocytes, keratinocytes and fibroblasts. Cells were harvested and kept at −80° C. until RNA isolation. Isolated RNA samples from Heart, Liver and Skeletal Muscle were purchased from commercial suppliers (Ambion) and kept at −80° C. until use in reverse transcription.


Analyzed Genes

10 genes were selected for the expression analysis considering the location of regions of differential methylation. In all of them, the region of differential methylation located in the promoter region, the table below under results provides the names of the analyzed genes.


Total RNA Isolation and RT-PCR:

Total RNA was isolated from cells and tissues using RNeasy kits (Qiagen, Hilden, Germany). Concentration and purity of the obtained RNA was determined spectrophotometrically, while the integrity was determined by electrophoresis on denaturing IM urea—2% agarose gel. cDNA was prepared using Omniscript RT kit (Qiagen, Hilden, Germany) with random hexamers in accordance to manufacturer's conditions. PCR was performed using 3 μl of the prepared cDNA, specific primers and HotStartTaq DNA polymerase kit in accordance with manufacturer's conditions. The PCR parameters used were: 15 min at 95° C. followed by 40 cycles of 1 min at 94° C., 1 min at the annealing temperature specific for each primer pair, and 1 min at 72° C., ending with a final extension for 10 min at 72° C. Primers were designed to bind in successive exons in order to avoid amplification in case contaminating genomic DNA was present. Amplification conditions for each fragment were determined experimentally by amplifying cDNA produced from Universal Human RNA (BioCat, Heidelberg, Germany), which is a pool total RNAs isolated from several tissues. The sequences of the specific primers for each analyzed gene are provided in the accompanying sequence listing according to the table below. It further shows the particular annealing temperature used in the amplification reaction. PCR products were separated through a 2.5% Agarose gel.


b) Results

All analyzed genes showed a correlation between high methylation and gene silencing. In all of them, the region of differential methylation was located in the promoter region. Table 10 shows the results for each analyzed gene. As an example the results for the three genes MYO18B, SLC22A1 and PLG are shown in FIG. 2. As a control the expression of SERPINB5 was also studied. Finally the expression of a housekeeping gene in all the studied samples demonstrated the feasibility of the assay and gave a baseline for the semiquantitative analysis of the expression.









TABLE 10







Analyzed genes, primers and annealling temperatures and results.















Differential








methylation


Annealing
Correlation


Gene
Gene SEQ
region
Forward Primer
Reverse Primer
temperature
methylation/


Name
ID NO:
position
SEQ ID NO:
SEQ ID NO:
(° C.)
expression
















RAET1E
127
Promoter
4945
4946
60
Inverse


PIB5PA
53
Promoter
4947
4948
60
Inverse


PLG
171
Promoter
4949
4950
60
Inverse


TGM3
180
Promoter
4951
4952
60
Inverse


SLC22A1
170
Promoter
4953
4954
60
Inverse


RP3-
151
Promoter
4955
4956
60
Inverse


398D13.4


OSM
39
Promoter
4957
4958
63
Inverse


MYO18B
12
Promoter
4959
4960
60
Inverse


CTA-
43
Promoter
4961
4962
60
Inverse


299D3.6


PARVG
85
Promoter
4963
4964
60
Inverse


SERPINB5

Promoter
4965
4966
60
Inverse








(Positive








control)


ACTB1


4967
4968
56
No correlation








(Negative








control)









Example 2
Large-Scale DNA Methylation Profiling of Human Chromosomes 6, 20 and 22

Using bisulfite DNA sequencing, we report high-resolution methylation reference profiles of human chromosomes 6, 20 and 22, providing a resource of about 1.9 million CpG measurements in 43 samples derived from 12 different (healthy) tissues.


It was the aim of the study to establish DNA methylation reference profiles for three human chromosomes from a number of healthy (no known disease phenotype) human tissues and primary cells. The study was further controlled for age and sex and comprised the analysis of 43 different samples derived from sperm, various primary cell types (dermal fibroblasts, dermal keratinocytes, dermal melanocytes, CD4+ and CD8+ lymphocytes) and tissues (heart muscle, skeletal muscle, liver and placenta, table 9). Tissues were pooled from up to three age- and sex-matched individuals (see table 9 for details). Primary cells were cultured for no more than three passages to minimize the risk of introducing aberrant methylation. Additionally, the methylation of selected amplicons were compared before and after cultivation with no difference in average methylation being detected. As dermal fibroblasts, keratinocytes and melanocytes are the major cell types constituting the human epidermis, we compared the average methylation of selected amplicons in these cell types with the corresponding values derived from additional human skin samples. No significant deviation between the methylation of the primary cells and tissues were detected, indicating that cell culturing for a limited number of passages does not change DNA methylation.


In total, we analyzed 2,524 amplicons associated with 873 genes on chromosomes 6, 20 and 22. Based on Ensembl (NCBI34) annotation, the amplicons were assigned to 6 distinct categories. Taking the number of biological and technical replicates into account, we have determined the methylation status of 1.88 million CpG sites. The corresponding data have been deposited into the public HEP database and can be accessed at www.epigenome.org.


Material and Methods

Cell and Tissue samples: Tissue samples were obtained from following sources: Asterand, (Detroit, US), Pathlore Plc. (Nottingham, UK), Tissue Transformation Technologies (T-cubed, Edison, US), Northwest Andrology (Missoula, US), NDRI (Philadelphia, US) and Biocat GmBH (Heidelberg, Germany). Only anonymized samples were used and ethical approval was obtained for the study. Contamination by blood cells is estimated to be low as blood specific methylation profiles were not detected in the tissues. Human primary cells were acquired from Cascade Biologics (Mansfield, United Kingdom), Cell Applications Inc. (San Diego, United States), Analytical Biological Services Inc. (Wilmington, US), Cambrex Bio Science (Verviers, Belgium) and from the DIGZ (Berlin, Germany). Dermal fibroblasts, keratinocytes and melanocytes were cultured according to the supplier's recommendations up to a maximum of 3 passages reducing the risk of aberrant methylation due to extended culturing. CD4+ T-lymphocytes were isolated from fresh whole blood by depletion of CD4+ monocytes followed by a negative selection. CD8+ cells were isolated from fresh whole blood by positive selection. Subsequent FACS analysis confirmed a purity of CD4+/CD8+ T-lymphocytes greater than 90%. In some cases, DNA samples were pooled according to the sex and age of the donors. All genders were confirmed by sex-specific PCR.


Amplicon selection and classification: Amplicons were selected and classified based on Ensembl (build NCBI 34) annotation, amplicons were designed in the following genomic regions:


5′-UTR: Overlapping by at least 200 bp with or within a core region of 2,000 bp upstream to 500 bp downstream of the TSS. Where multiple sites were annotated per gene, the first annotated TSS was used.


Exonic: Greater than 50% and at least 200 bp of amplicon overlapping with annotated exon. Intronic: Greater than 50% and at least 200 bp of amplicon overlapping with annotated intron. ECR (evolutionary conserved regions): ≧70% DNA sequence similarity (including ≧4 CpGs) for at least 100 bp between human and mouse non-coding sequences. Out of 3,249 ECRs identified on chromosome 20, 290 intergenic and 206 intronic (496 in total) ECRs were selected.


Sp1: Overlapping with putative Sp1 sites identified by ChIP-chip analysis.


ncRNA: CD box snoRNAs as described by Lestrade, L. and Weber, M. J. snoRNA-LBME-db, a comprehensive database of human H/ACA and C/D box snoRNAs. Nucleic Acids Res. 34, 158-162 (2006) and miRNAs as reported by Griffiths-Jones, S. The microRNA Registry. Nucleic Acids Res. 32, 109-111 (2004) located on chromosome 22.


Other: amplicons that are not located within a gene or a 5′-UTR and additionally do not belong to any other category. CGI (CpG island) were classified based on the criteria by Gardiner-Garden and Frommer J. Mol. Biol. 196, 261-282 (1987).


DNA extraction, PCR amplification and sequencing: DNA was extracted using the Qiagen DNA Genomic-tip kit according the manufacturer's recommendation. After quantification, DNA was bisulfite converted as previously described in PCT/WO/2005/038051 (2005). Bisulfite-specific primers with a minimum length of 18 bp were designed using a modified primer-3 program. The target sequence of the designed primers contained no CpGs allowing for an unbiased amplification of both hypo- and hypermethylated DNAs. All primers were tested for their ability to yield high quality sequences. Primers that gave rise to an amplicon of the expected size using non-bisulfite treated DNA as a template were discarded, thus ensuring the specificity for bisulphite-converted DNAs. Primers were also tested for specificity on bisulfite DNA by electronic PCR. DNA amplification was set up in 96-well plates using an automated pipeline. PCR amplicons were quality controlled by agarose gel electrophoresis, re-arrayed into 384-well plates for high-throughput processing, cleaned up using ExoSAP-IT (USB Corporation, Cleveland, Ohio) to remove any excess nucleotides and primers and sequenced directly in the forward and reverse directions. Sequencing was performed on ABI 3730 capillary sequencers using 1/32nd dilution of ABI Prism BigDye terminator V3.1 sequencing chemistry after hotstart (96° C. for 30 seconds) thermocycling (92° C. for 5 seconds, 50° C. for 5 seconds, 60° C. for 120 seconds×44 cycles) and ethanol precipitation. PCR fragments were sequenced using the same PCR amplification primers. PCR primers for the differentially methylated amplificates are provided in Table 4. Trace files and methylation signals at a given CpG site were quantified using the software ESME as previously described in Bioinformatics 20, 3005-3012 (2004).


The bisulfite sequencing-based approach chosen here allows one to measure DNA methylation with high reproducibility and accuracy, as independent measurements are derived from both the sense and antisense strands of a PCR amplicon (R=0.87; N=557,837). In addition, about 4.1% of the amplicons were subjected to independent PCR amplification and sequencing. These technical replicates also displayed high correlation (R=0.9; N=15,655). Furthermore, the signal is independent of the position of the measured CpG within the amplicon, which is supported by high correlation between measurements of the same CpGs in overlapping amplicons (R=0.85; N=91,528).


RNA extraction and RT-PCR. Aliquots of the same samples of the human melanocytes, keratinocytes, fibroblasts, CD4+ and CD8+ cells that were used for methylation analysis were used for RNA analysis. Primary cell cultures (maximum of 3 passages) of human melanocytes, keratinocytes and dermal fibroblasts cells were harvested and kept at −80° C. until RNA isolation. Isolated RNA samples from heart, liver and skeletal muscle were purchased from Ambion (Austin, US) and kept at −80° C. until used for reverse transcription. Total RNA was isolated using the RNeasy kit from Qiagen (Hilden, Germany) followed by cDNA synthesis using the Omniscript RT kit from the same supplier and random hexamers. PCR (92° C. for 1 minute, 55-63° C. (depending on assay) for 1 minute, 72° C. for 1 minute for 30 to 40 cycles (depending on assay)) was performed using the HotStartTaq DNA polymerase kit (Qiagen) with 3 μl of the prepared cDNA and gene-specific primers. All kits were used according to the manufacturer's recommendations. PCR products were analyzed by electrophoresis on 2.5% agarose gels. Universal RNA was obtained from Biocat (Heidelberg, Germany) and total RNA isolated from brain and sperm from Stratagene (La Jolla, Calif., US).


Analysis and Statistical methods: Methylation profiles were calculated as described previously in PLoS Biol. 2, 2170-2182 (2004) and are available from the HEP database/browser at www.epigenome.org. The methylation profile of each individual amplicon is provided in FIGS. 1.1 to 1.403.


Results

Each individual matrix represents the sequencing data for an individual amplificate. Each of the discrete blocks of the matrix represent a single sample type and are labeled ‘A’ through ‘L’, said letters representing in each case the following tissue/cell types: A: Melanocytes; B: Heart Muscle; C: Skeletal muscle; D: Liver; E: Sperm; F: Embryonic skeletal muscle; G: Embryonic liver; H: Placental; I: Fibroblast; J: Keratinocytes; K: CD8; L: CD4.


The SEQ ID NO: of the genomic region of each amplificate is shown to the left of the matrices. This may be cross referenced in Table 4 to determine the amplificate and primer sequences. Each row of a matrix represents a single CpG site within the amplificate (according to the corresponding SEQ ID NO: from Table 4) and is numbered accordingly, each column represents a single pooled DNA sample.


The degree of methylation is represented by the shade of each position within the column from black representing 100% methylation to light gray representing 0% methylation. White positions represented a measurement for which no data was available.


Differentially Methylated Regions (DMRS)

Kruskall-Wallis tests were used to determine differential methylation between tissues, measuring the proportion of uncorrected p-values that were smaller 0.001 for all CpGs. As this test is insensitive to samples that were only measured in a single sample such as sperm and placenta, the obtained number of DMRs is unlikely to be overstated due to putative aberrant methylation within these samples. Some DMRs were experimentally validated by sequencing independent DNA samples. Equality between two groups (age and sex) was performed using Wilcoxon tests.


Median CpG methylation values were used for the analysis of co-methylation. CpGs for which methylation values derived from both the forward and reverse strands displayed a difference of greater than 10% between the two values were excluded. Methylation changes were calculated based on the absolute methylation differences between CpG pairs of identical samples. To exclude a bias introduced by the amplicon selection, the analysis was performed using both, individual CpGs (window size 20,000 bp) and CpGs of the same amplicons. Co-methylation of CpGs was described as a function of the distance (in bp) displaying the observed ratio of similar methylation degree.


For scatter plots, equal amounts of measurements were binned and ranked by numerical order of the X-axis values, representing means of X− and Y− data. For box plots and histograms, data were binned according to the intervals indicated on the X-axis containing different numbers of measurements.


As a measure for the probability of differential methylation, amplicons were sorted by their p-value and binned by rank into groups of 200 and scanned using 211 vertebrate position-weight matrices from the TRANSFAC library40 (version 3.2). For each motif, we picked a threshold such that it matched around 40% of amplicons and performed a □2 test to determine if the hit rate of the motif varied significantly between the highest and lowest 200 amplicons, ranked by P-value of differential methylation.


Tables 6 and 7 provide an overview of the genomic regions and ranges of methylation within which the actual value of methylation of said genomic regions lies specific of the said cells, tissues and/or organs. The actual value of methylation is shown in FIGS. 1.1-1.403.


Table 11 provides an overview of 43 different samples derived from sperm, various primary cell types (dermal fibroblasts, dermal keratinocytes, dermal melanocytes, CD4+ and CD8+ lymphocytes) and tissues (heart muscle, skeletal muscle, liver and placenta. The study was controlled for age and sex and comprised the analysis. Tissues were pooled from up to three age- and sex-matched individuals.


Table 12 provides summary statistics.


Table 13 provides an overview of the most preferred of the genes or genomic regions and ranges of methylation within which the actual value of methylation of said genomic regions lies specific of the said cells, tissues and/or organs. The actual value of methylation is shown in FIGS. 1.1-1.403. We found 17% of the 873 analyzed genes differentially methylated in their 5′-untranslated regions (5′-UTR), in at least one of the tissues examined. Differential methylation is observed more frequently in evolutionary conserved regions (ECRs) than within 5′-UTRs, suggesting that methylation has a functional role beyond a direct effect on transcription via promoter methylation. About one third of the differentially methylated 5′-UTRs are inversely correlated with transcription of the respective mRNAs. We did not find any significant sex or age-dependent differences in our study, indicating that methylation is ontogenetically more stable than previously anticipated.













TABLE 11







Age




#
Tissue/cell type
(average)
Sex
Pool of







1
heart muscle
25 Y
female
3


2
heart muscle
23 Y
male
3


3
heart muscle
25 Y
male
3


4
heart muscle
78 Y
male
3


5
heart muscle
74 Y
male
3


6
heart muscle
62 Y
male
3


7
skeletal muscle
19 Y
male
3


8
skeletal muscle
30 Y
male
3


9
skeletal muscle
22 Y
male
3


10
skeletal muscle
59 Y
male
3


11
skeletal muscle
67 Y
male
3


12
skeletal muscle
74 Y
male
3


13
skeletal muscle
65 Y
female
3


14
skeletal muscle
67 Y
female
3


15
skeletal muscle
84 Y
female
2


16
liver
23 Y
male
3


17
liver
23 Y
male
3


18
liver
30 Y
male
3


19
liver
82 Y
male
3


20
liver
65 Y
male
3


21
liver
74 Y
male
3


22
liver
70 Y
female
3


23
liver
81 Y
female
3


24
sperm
24 Y
male
3


25
fetal skeletal muscle
23 W
male
3


26
fetal liver
24 W
male
3


27
placenta
NB
female
3


28
melanocytes
30 Y
female
1


29
melanocytes
41 Y
female
1


30
melanocytes
42 Y
female
1


31
dermal fibroblasts
43 Y
female
2


32
dermal fibroblasts
41 Y
female
2


33
dermal fibroblasts
39 Y
female
2


34
dermal keratinocytes
43 Y
female
3


35
dermal keratinocytes
39 Y
female
3


36
dermal keratinocytes
33 Y
female
2


37
CD4 lymphocytes
20 Y
male
1


38
CD8 lymphocytes
59 Y
male
1


39
CD8 lymphocytes
60/61 Y
female
1


40
CD8 lymphocytes
62 Y
male
1


41
CD4 lymphocytes
59/60 Y
female
1


42
CD4 lymphocytes
61 Y
female
1


43
CD4 lymphocytes
62 Y
female
1









Table 11 provides an overview of 43 different samples derived from sperm, various primary cell types (dermal fibroblasts, dermal keratinocytes, dermal melanocytes, CD4+ and CD8+ lymphocytes) and tissues (heart muscle, skeletal muscle, liver and placenta.














TABLE 12








Chromosome
Chromosome
Chromosome



Total
6
20
22




















CpG islands on chromosome
2,279
1,07
662
547


CpG islands covered
511
256
29
226


CpG islands percentage covered
22%
24%
4%
41%


Genes covered
873
383
89
401


Exons covered
853
454
23
376


Introns covered
920
465
118
337


Number of tissues analyzed
12


Number of samples analyzed
43


Average length of amplicon +/− SD
411 +/−



77 bp


Average number of CpGs per
16 +/− 10.8


amplicon


Total number of different amplicons
2,524


Number of CpGs analyzed
1,885,003









Table 12 provides summary statistics.














TABLE 13







Gene
Ensembl ID
CD4 T-lymphocytes
CD8 T-lymphocytes
Embryonic liver
Em. ske. muscle





ZNRF3
ENSG00000183579
90
90
80
30


SLC7A4
OTTHUMG00000030129
0
0
0
0


Myosin-18B
ENSG00000133454
100
90
80
40


(MYO18B)


Glycoprotein Ib
OTTHUMT00000075045
10
10
10
70


(platelet), beta


polypeptide


RP1-47A17.8
OTTHUMG00000030878
80
80
70
20


Oncostatin M
ENSG00000099985
0
0
40
90


(OSM)


CTA-299D3.6
OTTHUMG00000030140
90
90
80
70


CTA-941F9.6
OTTHUMG00000030231
90
90
90
80


Cytohesin-4
ENSG00000100055
0
0
30
50


PIB5PA
ENSG00000185133
60
70
60
10


SUSD2
ENSG00000099994
100
90
80
40


No gene associated
NA
90
90
80
30


RP3-438O4.2
OTTHUMG00000030922
90
90
90
30


RP4-756G23.1
OTTHUMG00000030852
90
90
80
50


Somatostatin
ENSG00000183473
90
90
80
30


receptor type 3


(SSTR3)


Bcl-2 interacting
ENSG00000100290
20
40
40
50


killer (BIK)


GAS2L1
ENSG00000185340
90
90
90
80


RP3-355C18.2
OTTHUMG00000030072
60
60
80
70


Gamma-parvin
ENSG00000138964
30
20
70
90


CARMA 3
ENSG00000100065
100
100
90
NA


TMPRSS6
ENSG00000187045
10
30
50
0


Platelet-derived
ENSG00000100311
20
20
20
0


growth factor B


chain precursor


(PDGFB)


CELSR1 Cadherin
ENSG00000075275
10
10
40
60


T-box transcription
ENSG00000184058
10
10
NA
40


factor (TBX1)


Q6ZRW2_HUMAN
ENSG00000178199
40
30
70
30


NKG2DL4
ENSG00000164520
70
90
70
70


(RAET1E)


DAAM2
ENSG00000146122
90
90
90
60


RP1-47M23.1
OTTHUMG00000015313
10
0
20
40


RP11-397G17.1
OTTHUMG00000014829
90
90
80
90


Nesprin-1 (Syne-1)
ENSG00000131018
100
100
70
NA


Myogenic repressor
ENSG00000112559
80
80
50
50


I-mf (MDFI)


RP11-174C7.4
OTTHUMG00000015553
80
80
80
40


CMAH
OTTHUMG00000016099
90
100
90
80


PKHD1
ENSG00000170927
90
80
70
50


glutathione
OTTHUMG00000016307
20
10
30
80


peroxidase 5


(GPX5)


GRIK2
ENSG00000164418
10
20
10
10


SLC22A1
ENSG00000112499
100
100
100
90


RP11-235G24.1
OTTHUMG00000015959
90
90
80
90


TBX18
ENSG00000112837
10
20
0
40


TGM3
ENSG00000125780
90
80
70
NA


RIN2
OTTHUMG00000031996
30
50
40
20


SLC24A3
OTTHUMG00000031993
100
100
100
60


Q9ULE8_HUMAN
ENSG00000188559
100
100
80
NA


C20orf117
OTTHUMG00000032395
80
90
70
10


Breast carcinoma
ENSG00000124243
10
10
10
20


amplified sequence


4 (BCAS4)


nuclear factor of
OTTHUMG00000032747
100
90
90
40


activated T-cells


(NFATC2)


SCUBE1
ENSG00000159307
10
10
0
40




















Gene
Fibroblasts
Heart muscle
Keratinocytes
Liver
Melanocytes
Placenta
Skelatal muscle
Sperm







ZNRF3
80
70
90
90
90
60
30
100



SLC7A4
0
0
10
10
50
10
0
0



Myosin-18B
90
60
90
90
90
50
40
NA



(MYO18B)



Glycoprotein Ib
90
20
40
10
50
70
80
0



(platelet), beta



polypeptide



RP1-47A17.8
20
80
90
80
70
70
60
90



Oncostatin M
90
90
100
90
100
90
90
90



(OSM)



CTA-299D3.6
70
70
90
80
30
70
70
100



CTA-941F9.6
90
90
10
80
90
90
80
90



Cytohesin-4
50
50
10
50
60
30
50
90



PIB5PA
10
10
10
20
10
10
10
60



SUSD2
50
80
40
60
50
60
50
90



No gene associated
10
40
10
60
60
20
20
90



RP3-438O4.2
10
70
20
80
20
20
20
90



RP4-756G23.1
50
80
80
90
20
70
50
90



Somatostatin
10
20
40
30
20
20
20
90



receptor type 3



(SSTR3)



Bcl-2 interacting
80
80
90
80
80
80
50
90



killer (BIK)



GAS2L1
80
80
10
90
90
90
70
20



RP3-355C18.2
60
80
10
70
80
50
50
100



Gamma-parvin
90
90
90
80
90
80
90
100



CARMA 3
80
90
80
100
90
80
50
100



TMPRSS6
10
30
60
80
10
30
0
0



Platelet-derived
50
30
10
70
70
20
20
0



growth factor B



chain precursor



(PDGFB)



CELSR1 Cadherin
70
70
80
80
80
80
60
0



T-box transcription
80
10
0
20
70
80
70
NA



factor (TBX1)



Q6ZRW2_HUMAN
30
60
20
80
40
60
20
100



NKG2DL4
50
80
0
100
70
50
50
100



(RAET1E)



DAAM2
90
90
90
20
90
90
80
100



RP1-47M23.1
40
80
70
40
60
60
60
90



RP11-397G17.1
80
90
20
50
80
70
80
90



Nesprin-1 (Syne-1)
10
20
10
40
0
60
20
90



Myogenic repressor
80
40
0
50
60
50
80
0



I-mf (MDFI)



RP11-174C7.4
0
50
70
80
50
40
30
80



CMAH
90
90
0
90
100
90
80
0



PKHD1
40
50
10
40
30
50
60
100



glutathione
40
50
90
70
40
80
70
100



peroxidase 5



(GPX5)



GRIK2
80
20
90
10
70
50
10
10



SLC22A1
100
100
100
60
100
90
100
100



RP11-235G24.1
50
80
90
60
70
80
90
80



TBX18
90
30
10
20
60
40
60
10



TGM3
70
70
30
90
80
50
40
90



RIN2
20
20
90
90
20
30
10
NA



SLC24A3
90
80
90
90
90
NA
50
NA



Q9ULE8_HUMAN
90
100
0
60
100
90
100
100



C20orf117
0
30
10
50
10
20
40
10



Breast carcinoma
20
20
10
20
70
20
30
80



amplified sequence



4 (BCAS4)



nuclear factor of
10
70
10
50
10
40
40
100



activated T-cells



(NFATC2)



SCUBE1
70
60
50
20
80
60
70
70










Table 13 provides an overview of the most preferred of the genes or genomic regions and ranges of methylation within which the actual value of methylation of said genomic regions lies specific of the said cells, tissues and/or organs. The actual value of methylation is shown in FIGS. 1.1-1.403.


LITERATURE



  • Murakami M, Masuda S, Shimbara S, Ishikawa Y, Ishii T, Kudo I. Cellular distribution, post-translational modification, and tumorigenic potential of human group III secreted phospholipase A(2). J Biol Chem. 2005 Jul. 1; 280(26):24987-98. Epub 2005 Apr. 29.

  • Habib S J, Waizenegger T, Lech M, Neupert W, Rapaport D. Assembly of the TOB complex of mitochondria. J Biol Chem. 2005 Feb. 25; 280(8):6434-40. Epub 2004 Dec. 7.

  • Igarashi P, Shao X, McNally B T, Hiesberger T. Roles of HNF-1beta in kidney development and congenital cystic diseases. Kidney Int. 2005 November; 68(5):1944-7.

  • Bergmann C, Kupper F, Domia C, Schneider F, Senderek J, Zerres K. Algorithm for efficient PKHD1 mutation screening in autosomal recessive polycystic kidney disease (ARPKD). Hum Mutat. 2005 March; 25(3):225-31.

  • Menezes L F, Cai Y, Nagasawa Y, Silva A M, Watkins M L, Da Silva A M, Somlo S, Guay-Woodford L M, Germino G G, Onuchic L F. Polyductin, the PKHD1 gene product, comprises isoforms expressed in plasma membrane, primary cilium, and cytoplasm. Kidney Int. 2004 October; 66(4):1345-55.

  • Gresh L, Fischer E, Reimann A, Tanguy M, Garbay S, Shao X, Hiesberger T, Fiette L, Igarashi P, Yaniv M, Pontoglio M. A transcriptional network in polycystic kidney disease. EMBO J. 2004 Apr. 7; 23(7): 1657-68. Epub 2004 Mar. 18.

  • Zhang M Z, Mai W, Li C, Cho S Y, Hao C, Moeckel G, Zhao R, Kim I, Wang J, Xiong H, Wang H, Sato Y, Wu Y, Nakanuma Y, Lilova M, Pei Y, Harris R C, Li S, Coffey R J, Sun L, Wu D, Chen X Z, Breyer M D, Zhao Z J, McKanna J A, Wu G. PKHD1 protein encoded by the gene for autosomal recessive polycystic kidney disease associates with basal bodies and primary cilia in renal epithelial cells. Proc Natl Acad Sci USA. 2004 Feb. 24; 101(8):2311-6.

  • Onuchic L F, Furu L, Nagasawa Y, Hou X, Eggermann T, Ren Z, Bergmann C, Senderek J, Esquivel E, Zeltner R, Rudnik-Schoneborn S, Mrug M, Sweeney W, Avner E D, Zerres K, Guay-Woodford L M, Somlo S, Germino G G. PKHD1, the polycystic kidney and hepatic disease I gene, encodes a novel large protein containing multiple immunoglobulin-like plexin-transcription-factor domains and parallel beta-helix I repeats. Am J Hum Genet. 2002 May; 70(5):1305-17. Epub 2002 Mar. 15.

  • McCann J A, Xu Y Q, Frechette R, Guazzarotti L, Polychronakos C. The insulin-like growth factor-II receptor gene is associated with type I diabetes: evidence of a maternal effect. J Clin Endocrinol Metab. 2004 November; 89(11):5700-6. PMID: 15531531

  • Grundemann, D.; Gorboulev, V.; Gambaryan, S.; Veyhl, M.; Koepsell, H. Drug excretion mediated by a new prototype of polyspecific transporter. Nature 372: 549-552, 1994.

  • Leabman, M. K.; Huang, C. C.; DeYoung, J.; Carlson, E. J.; Taylor, T. R.; de la Cruz, M.; Johns, S. J.; Stryke, D.; Kawamoto, M.; Urban, T. J.; Kroetz, D. L.; Ferrin, T. E.: Clark, A. G.; Risch, N.; Herskowitz, I.; Giacomini, K. M.; {Pharmacogenetics of Membrane Transporters Investigators}: Natural variation in human membrane transporter genes reveals evolutionary and functional constraints. Proc. Nat. Acad. Sci. 100: 5896-5901, 2003.

  • Shu, Y.; Leabman, M. K.; Feng, B.; Mangravite, L. M.; Huang, C. C.; Stryke, D.; Kawamoto, M.; Johns, S. J.; DeYoung, J.; Carlson, E.; Ferrin, T. E.; Herskowitz, 1.; Giacomini, K. M.; Pharmacogenetics of Membrane Transporters Investigators: Evolutionary conservation predicts function of variants of the human organic cation transporter, OCT1. Proc. Nat. Acad. Sci. 100: 5902-5907, 2003.

  • Bourdet D L, Pritchard J B, Thakker D R. Differential substrate and inhibitory activities of ranitidine and famotidine toward human organic cation transporter 1 (hOCT1; SLC22A1), hOCT2 (SLC22A2), and hOCT3 (SLC22A3). J Pharmacol Exp Ther. 2005 December; 315(3):1288-97

  • Lips K S, Volk C, Schmitt B M, Pfeil U, Arndt P, Miska D, Ermert L, Kummer W, Koepsell H. Polyspecific cation transporters mediate luminal release of acetylcholine from bronchial epithelium. Am J Respir Cell Mol Biol. 2005 July; 33(1):79-88.)

  • Grover B, Buckley D, Buckley A R, Cacini W. Reduced expression of organic cation transporters ROCT1 and rOCT2 in experimental diabetes. J Pharmacol Exp Ther. 2004 March; 308(3):949-56.)

  • Wang D S, Kusuhara H, Kato Y, Jonker J W, Schinkel A H, Sugiyama Y. Involvement of organic cation transporter I in the lactic acidosis caused by metformin. Mol Pharmacol. 2003 April; 63(4):844-8.)

  • Sleutels F, Zwart R, Barlow D P. The non-coding Air RNA is required for silencing autosomal imprinted genes. Nature. 2002 Feb. 14; 415(6873):810-3.

  • Grover B, Buckley D, Buckley A R, Cacini W. Reduced expression of organic cation transporters rOCT1 and rOCT2 in experimental diabetes. J Pharmacol Exp Ther. 2004 March; 308(3):949-56. Epub 2004 Jan. 8.

  • Karbach U, Kricke J, Meyer-Wentrup F, Gorboulev V, Volk C, Loffing-Cueni D, Kaissling B, Bachmann S, Koepsell H. Localization of organic cation transporters OCT1 and OCT2 in rat kidney. Am J Physiol Renal Physiol. 2000 October; 279(4):F679-87.

  • Endoh M, Tamura G, Honda T, Homma N, Terashima M, Nishizuka S, Motoyama T. RASSF2, a potential tumor suppressor, is silenced by CpG island hypermethylation in gastric cancer. Br J Cancer. 2005 Dec. 12; 93(12):1395-1399.

  • Akino K, Toyota M, Suzuki H, Mita H, Sasaki Y, Ohe-Toyota M, Issa J P, Hinoda Y, Imai K, Tokino T. The Ras effector RASSF2 is a novel tumor-suppressor gene in human colorectal cancer. Gastroenterology. 2005 July; 129(1):156-69.

  • Hesson L B, Wilson R, Morton D, Adams C, Walker M, Maher E R, Latif F. CpG island promoter hypermethylation of a novel Ras-effector gene RASSF2A is an early event in colon carcinogenesis and correlates inversely with K-ras mutations. Oncogene. 2005 Jun. 2; 24(24):3987-94.

  • Vos M D, Ellis C A, Elam C, Ulku A S, Taylor B J, Clark G J. RASSF2 is a novel K-Ras-specific effector and potential tumor suppressor. J Biol Chem. 2003 Jul. 25; 278(30):28045-51. Epub 2003 May 5. PMID: 12732644

  • Ooms L M, Fedele C G, Astle M V, Ivetac I, Cheung V, Pearson R B, Layton M J, Forrai A, Nandurkar H H, Mitchell C A. The Inositol Polyphosphate 5-Phosphatase, PIPP, Is a Novel Regulator of Phosphoinositide 3-Kinase-dependent Neurite Elongation. Mol Biol Cell 2005 Nov. 9;

  • Ohkura S, Kondoh N, Hada A, Arai M, Yamazaki Y, Sindoh M, Takahashi M, Matsumoto I, Yamamoto M. Differential expression of the keratin-4, -13, -14, -17 and transglutaminase 3 genes during the development of oral squamous cell carcinoma from leukoplakia. Oral Oncol. 2005 July; 41(6):607-13. Epub 2005 Apr. 14.

  • Zhang J, Zhi H Y, Ding F, Luo A P, Liu Z H. Transglutaminase 3 expression in C57BL/6J mouse embryo epidermis and the correlation with its differentiation. Cell Res. 2005 February; 15(2): 105-10.

  • Zainelli G M, Dudek N L, Ross C A, Kim S Y, Muma N A. Mutant huntingtin protein: a substrate for transglutaminase 1, 2, and 3. J Neuropathol Exp Neurol. 2005 January; 64(1):58-65.

  • Raghunath M, Tontsidou L, Oji V, Aufenvenne K, Schurmeyer-Horst F, Jayakumar A, Stander H, Smolle J, Clayman G L, Traupe H. SPINK5 and Netherton syndrome: novel mutations, demonstration of missing LEKTI, and differential expression of transglutaminases. J Invest Dermatol. 2004 September; 123(3):474-83.

  • Luo A, Kong J, Hu G, Liew C C, Xiong M, Wang X, Ji J, Wang T, Zhi H, Wu M, Liu Z. Discovery of Ca2+-relevant and differentiation-associated genes downregulated in esophageal squamous cell carcinoma using cDNA microarray. Oncogene. 2004 Feb. 12; 23(6):1291-9.

  • Gonzalez H E, Gujrati M, Frederick M, Henderson Y, Arumugam J, Spring P W, Mitsudo K, Kim H W, Clayman G L. Identification of 9 genes differentially expressed in head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg. 2003 July; 129(7):754-9.

  • Hitomi K, Presland R B, Nakayama T, Fleckman P, Dale B A, Maki M. Analysis of epidermal-type transglutaminase (transglutaminase 3) in human stratified epithelia and cultured keratinocytes using monoclonal antibodies. J Dermatol Sci. 2003 August; 32(2):95-103.

  • Nakano T, Tani M, Nishioka M, Kohno T, Otsuka A, Ohwada S, Yokota J. Genetic and epigenetic alterations of the candidate tumor-suppressor gene MYO18B, on chromosome arm 22q, in colorectal cancer. Genes Chromosomes Cancer. 2005 June; 43(2):162-71.

  • Yanaihara N, Nishioka M, Kohno T, Otsuka A, Okamoto A, Ochial K, Tanaka T, Yokota J. Reduced expression of MYO18B, a candidate tumor-suppressor gene on chromosome arm 22q, in ovarian cancer. Int J Cancer. 2004 Oct. 20; 112(1):150-4.

  • Nishioka M, Kohno T, Tani M, Yanaihara N, Tomizawa Y, Otsuka A, Sasaki S, Kobayashi K, Niki T, Maeshima A, Sekido Y, Minna J D, Sone S, Yokota J. MYO18B, a candidate tumor suppressor gene at chromosome 22q12.1, deleted, mutated, and methylated in human lung cancer. Proc Natl Acad Sci USA. 2002 Sep. 17; 99(19):12269-74. Epub 2002 Sep. 3.

  • Kumar U, Grigorakis S I, Watt H L, Sasi R, Snell L, Watson P, Chaudhari S. Somatostatin receptors in primary human breast cancer: quantitative analysis of mRNA for subtypes 1-5 and correlation with receptor protein expression and tumor pathology. Breast Cancer Res Treat. 2005 July; 92(2): 175-86.

  • Kumar U. Expression of somatostatin receptor subtypes (SSTR1-5) in Alzheimer's disease brain: an immunohistochemical analysis. Neuroscience. 2005; 134(2):525-38.

  • Liu H L, Huo L, Wang L. Octreotide inhibits proliferation and induces apoptosis of hepatocellular carcinoma cells. Acta Pharmacol Sin. 2004 October; 25(10):1380-6.

  • Hu C, Yi C, Hao Z, Cao S, Li H, Shao X, Zhang J, Qiao T, Fan D. The effect of somatostatin and SSTR3 on proliferation and apoptosis of gastric cancer cells. Cancer Biol Ther. 2004 August; 3(8):726-30. Epub 2004 Aug. 10.

  • Goodyer C G, Grigorakis S I, Patel Y C, Kumar U. Developmental changes in the expression of somatostatin receptors (1-5) in the brain, hypothalamus, pituitary and spinal cord of the human fetus. Neuroscience. 2004; 125(2):441-8.

  • Jia W D, Xu G L, Xu R N, Sun H C, Wang L, Yu J H, Wang J, Li J S, Zhai Z M, Xue Q. Octreotide acts as an antitumor angiogenesis compound and suppresses tumor growth in nude mice bearing human hepatocellular carcinoma xenografts. J Cancer Res Clin Oncol. 2003 June; 129(6):327-34. Epub 2003 Jun. 14.

  • Vikic-Topic S, Raisch K P, Kvols L K, Vuk-Pavlovic S. Expression of somatostatin receptor subtypes in breast carcinoma, carcinoid tumor, and renal cell carcinoma. J Clin Endocrinol Metab. 1995 October; 80(10):2974-9.

  • Drebin J A, Hartzell S W, Griffin C, Campbell M J, Niederhuber J E. Molecular cloning and chromosomal localization of the human homologue of a B-lymphocyte specific protein tyrosine kinase (blk). Oncogene. 1995 Feb. 2; 10(3):477-86.

  • Lin Y H, Shin E J, Campbell M J, Niederhuber J E. Transcription of the blk gene in human B lymphocytes is controlled by two promoters. J Biol Chem. 1995 Oct. 27; 270(43):25968-75.

  • Law D A, Gold M R, DeFranco A L. Examination of B lymphoid cell lines for membrane immunoglobulin-stimulated tyrosine phosphorylation and src-family tyrosine kinase mRNA expression. Mol Immunol. 1992 July-August; 29(7-8):917-26.

  • Yao X R, Scott D W. Antisense oligodeoxynucleotides to the blk tyrosine kinase prevent anti-mu-chain-mediated growth inhibition and apoptosis in a B-cell lymphoma. Proc Natl Acad Sci USA. 1993 Sep. 1; 90(17):7946-50.

  • Vallat L, Magdelenat H, Merle-Beral H, Masdehors P, Potocki de Montalk G, Davi F, Kruhoffer M, Sabatier L, Orntoft T F, Delic J. The resistance of B-CLL cells to DNA damage-induced apoptosis defined by DNA microarrays. Blood. 2003 Jun. 1; 101(11):4598-606. Epub 2003 Feb. 13.

  • Texido G, Su I H, Mecklenbrauker I, Saijo K, Malek S N, Desiderio S, Rajewsky K, Tarakhovsky A. The B-cell-specific Src-family kinase Blk is dispensable for B-cell development and activation. Mol Cell Biol. 2000 February; 20(4):1227-33.

  • Malek S N, Dordai D I, Reim J, Dintzis H, Desiderio S. Malignant transformation of early lymphoid progenitors in mice expressing an activated Blk tyrosine kinase. Proc Natl Acad Sci USA. 1998 Jun. 23; 95(13):7351-6.

  • Goriounov D, Leung C L, Liem R K. Protein products of human Gas2-related genes on chromosomes 17 and 22 (hGAR17 and hGAR22) associate with both microfilaments and microtubules. J Cell Sci. 2003 Mar. 15; 116(Pt 6):1045-58. PMID: 12584248

  • Brancolini C, Bottega S, Schneider C. Gas2, a growth arrest-specific protein, is a component of the microfilament network system. J Cell Biol. 1992 June; 117(6):1251-61. PMID: 1607387

  • Stilo R, Liguoro D, Di Jeso B, Formisano S, Consiglio E, Leonardi A, Vito P. Physical and functional interaction of CARMA1 and CARMA3 with Ikappa kinase gamma-NFkappaB essential modulator. J Biol Chem. 2004 Aug. 13; 279(33):34323-31. Epub 2004 Jun. 7.

  • Rauch A, Devriendt K, Koch A, Rauch R, Gewillig M, Kraus C, Weyand M, Singer H, Reis A, Hofbeck M. Assessment of association between variants and haplotypes of the remaining TBX1 gene and manifestations of congenital heart defects in 22q11.2 deletion patients. J Med Genet. 2004 April; 41(4):e40. No abstract available.

  • Vitelli F, Taddei I, Morishima M, Meyers E N, Lindsay E A, Baldini A. A genetic link between Tbx1 and fibroblast growth factor signaling. Development. 2002 October; 129(19):4605-11. PMID: 12223416

  • Zhang L, Zhong T, Wang Y, Jiang Q, Song H, Gui Y. TBX1, a DiGeorge syndrome candidate gene, is inhibited by retinoic acid. Int J Dev Biol. 2006; 50(1):55-61. Nakaya M A, Habas R, Biris K, Dunty W C Jr, Kato Y, He X, Yamaguchi T P. Identification and comparative expression analyses of Daam genes in mouse and Xenopus. Gene Expr Patterns. 2004 November; 5(1):97-105.

  • Kida Y, Shiraishi T, Ogura T. Identification of chick and mouse Daam1 and Daam2 genes and their expression patterns in the central nervous system. Brain Res Dev Brain Res. 2004 Oct. 15; 153(1):143-50.

  • Busso D, Cohen D J, Hayashi M, Kasahara M, Cuasnicu P S. Human testicular protein TPX1/CRISP-localization in spermatozoa, fate after capacitation and relevance for gamete interaction. Mol Hum Reprod. 2005 April; 11(4):299-305. Epub 2005 Feb. 25.

  • Wang H, Zhou Z, Xu M, Li J, Xiao J, Xu Z Y, Sha J. A spermatogenesis-related gene expression profile in human spermatozoa and its potential clinical applications. J Mol Med. 2004 May; 82(5):317-24. Epub 2004 Feb. 24.

  • Haendler B, Toda I, Sullivan D A, Schleuning W D. Expression of transcripts for cysteine-rich secretory proteins (CRISPs) in the murine lacrimal gland. J Cell Physiol. 1999 March; 178(3):371-8.

  • Grady R M, Starr D A, Ackerman G L, Sanes J R, Han M. Syne proteins anchor muscle nuclei at the neuromuscular junction. Proc Natl Acad Sci USA. 2005 Mar. 22; 102(12):4359-64. Epub 2005 Mar. 4.

  • Pare G C, Easlick J L, Mislow J M, McNally E M, Kapiloff M S. Nesprin-1alpha contributes to the targeting of mAKAP to the cardiac myocyte nuclear envelope. Exp Cell Res. 2005 Feb. 15; 303(2):388-99.

  • Fan J, Beck K A. A role for the spectrin superfamily member Syne-1 and kinesin II in cytokinesis. J Cell Sci. 2004 Feb. 1; 117(Pt 4):619-29. Epub 2004 Jan. 6.

  • Gough L L, Fan J, Chu S, Winnick S, Beck K A. Golgi localization of Syne-1. Mol Biol Cell. 2003 June; 14(6):2410-24. Epub 2003 Mar. 7.

  • Zhang Q, Skepper J N, Yang F, Davies J D, Hegyi L, Roberts R G, Weissberg P L, Ellis J A, Shanahan C M. Nesprins: a novel family of spectrin-repeat-containing proteins that localize to the nuclear membrane in multiple tissues. J Cell Sci. 2001 December; 114(Pt 24):4485-98.

  • Kusano S, Raab-Traub N. I-mfa domain proteins interact with Axin and affect its regulation of the Wnt and c-Jun N-terminal kinase signaling pathways. Mol Cell Biol. 2002 September; 22(18):6393-405.

  • Tsuji K, Kraut N, Groudine M, Noda M. Vitamin D(3) enhances the expression of 1-mfa, an inhibitor of the MyoD family, in osteoblasts. Biochim Biophys Acta. 2001 May 28; 1539(1-2):122-30. Kraut N, Snider L, Chen C M, Tapscott S J, Groudine M. Requirement of the mouse 1-mfa gene for placental development and skeletal patterning. EMBO J. 1998 Nov. 2; 17(21):6276-88. Hall L, Williams K, Perry A C, Frayne J, Jury J A. The majority of human glutathione peroxidase type 5 (GPX5) transcripts are incorrectly spliced: implications for the role of GPX5 in the male reproductive tract. Biochem J. 1998 Jul. 1; 333 (Pt 1):5-9. PMID: 9639555

  • Drevet J R, Lareyre J J, Schwaab V, Vernet P, Dufaure J P. The PEA3 protein of the Ets oncogene family is a putative transcriptional modulator of the mouse epididymis-specific glutathione peroxidase gene gpx5. Mol Reprod Dev. 1998 February; 49(2):131-40.

  • Lareyre J J, Claessens F, Rombauts W, Dufaure J P, Drevet J R. Characterization of an androgen response element within the promoter of the epididymis-specific murine glutathione peroxidase 5 gene. Mol Cell Endocrinol. 1997 Apr. 25; 129(1):33-46.

  • Karjalainen J M, Kellokoski J K, Mannermaa A J, Kujala H E, Moisio K I, Mitchell P J, Eskelinen M J, Alhava E M, Kosma V M. Failure in post-transcriptional processing is a possible inactivation mechanism of AP-2alpha in cutaneous melanoma. Br J Cancer. 2000 June; 82(12):2015-21. PMID: 10864211

  • Luscher B, Mitchell P J, Williams T, Tjian R. Regulation of transcription factor AP-2 by the morphogen retinoic acid and by second messengers. Genes Dev. 1989 October; 3(10):1507-17.

  • Melki S A, Abumrad N A. Expression of the adipocyte fatty acid-binding protein in streptozotocin-diabetes: effects of insulin deficiency and supplementation. J Lipid Res. 1993 September; 34(9): 1527-34.

  • Turner B C, Zhang J, Gumbs A A, Maher M G, Kaplan L, Carter D, Glazer P M, Hurst H C, Haffly B G, Williams T. Expression of AP-2 transcription factors in human breast cancer correlates with the regulation of multiple growth factor signalling pathways. Cancer Res. 1998 Dec. 1; 58(23):5466-72.

  • Linton M F, Fazio S. Macrophages, inflammation, and atherosclerosis. Int J Obes Relat Metab Disord. 2003 December; 27 Suppl S35-40. Review.

  • Jean D, Gershenwald J E, Huang S, Luca M, Hudson M J, Tainsky M A, Bar-Eli M. Loss of AP-2 results in up-regulation of MCAM/MUC18 and an increase in tumor growth and metastasis of human melanoma cells. J Biol Chem. 1998 Jun. 26; 273(26):16501-8. PMID: 9632718

  • Ropponen K M, Kellokoski J K, Pirinen R T, Moisio K I, Eskelinen M J, Alhava E M, Kosma V M. Expression of transcription factor AP-2 in colorectal adenomas and adenocarcinomas; comparison of immunohistochemistry and in situ hybridization. J Clin Pathol. 2001 July; 54(7):533-8. PMID: 11429425

  • Zhang L, Zhan S, Navid F, Li Q, Choi Y H, Kim M, Seth P, Helman L J. AP-2 may contribute to IGF-II overexpression in rhabdomyosarcoma. Oncogene. 1998 Sep. 10; 17(10):1261-70. PMID: 9771969

  • Zhang Q, Rubenstein J N, Jang T L, Pins M, Javonovic B, Yang X, Kim S J, Park I, Lee C. Insensitivity to transforming growth factor-beta results from promoter methylation of cognate receptors in human prostate cancer cells (LNCaP). Mol Endocrinol. 2005 September; 19(9):2390-9. Epub 2005 May 19. PMID: 15905358 [PubMed—in process]

  • Harder A, Rosche M, Reuss D E, Holtkamp N, Uhlmann K, Friedrich R, Mautner V F, von Deimling A. Methylation analysis of the neurofibromatosis type 1 (NF1) promoter in peripheral nerve sheath tumors. Eur J Cancer. 2004 December; 40(18):2820-8. PMID: 15571966

  • Zhu C H, Huang Y, Broman M T, Domann F E. Expression of AP-2 alpha in SV40 immortalized human lung fibroblasts is associated with a distinct pattern of cytosine methylation in the AP-2 alpha promoter. Biochim Biophys Acta. 2001 May 28; 1519(1-2):85-91. PMID: 11406275

  • Sergeeva O A, Amberger B T, Eriksson K S, Scherer A, Haas H L. Co-ordinated expression of 5-HT2C receptors with the NCX1 Na+/Ca2+ exchanger in histaminergic neurones. J Neurochem. 2003 November; 87(3):657-64. PMID: 14535948

  • Kraev A, Quednau B D, Leach S, Li X F, Dong H, Winkfein R, Perizzolo M, Cai X, Yang R, Philipson K D, Lytton J. Molecular cloning of a third member of the potassium-dependent sodium-calcium exchanger gene family, NCKX3. J Biol Chem. 2001 Jun. 22; 276(25):23161-72. Epub 2001 Apr. 9. PMID: 11294880

  • Barlund M, Monni O, Weaver J D, Kauraniemi P, Sauter G, Heiskanen M, Kallioniemi O P, Kallioniemi A. Cloning of BCAS3 (17q23) and BCAS4 (20q13) genes that undergo amplification, overexpression, and fusion in breast cancer. Genes Chromosomes Cancer. 2002 December; 35(4):311-7. PMID: 12378525

  • Feske S, Draeger R, Peter H H, Eichmann K, Rao A. The duration of nuclear residence of NFAT determines the pattern of cytokine expression in human SCID T cells. J Immunol. 2000 Jul. 1; 165(1):297-305. PMID: 10861065

  • Amasaki Y, Miyatake S, Arai N, Arai K. Regulation of nuclear factor of activated T-cell family transcription factors during T-cell development in the thymus. J Allergy Clin Immunol. 2000 July; 106(1 Pt 2): S1-9. PMID: 10887328

  • Avni O, Lee D, Macian F, Szabo S J, Glimcher L H, Rao A. T(H) cell differentiation is accompanied by dynamic changes in histone acetylation of cytokine genes. Nat Immunol. 2002 July; 3(7):643-51. Epub 2002 Jun. 10. PMID: 12055628

  • Willard-Gallo K E, Badran B M, Ravoet M, Zerghe A, Burny A, Martiat P, Goldman M, Roufosse F, Sibille C. Defective CD3gamma gene transcription is associated with NFATc2 overexpression in the lymphocytic variant of hypereosinophilic syndrome. Exp Hematol. 2005 October; 33(10):1147-59. PMID: 16219537

  • Yang R B, Ng C K, Wasserman S M, Colman S D, Shenoy S, Mehraban F, Komuves L G, Tomlinson J E, Topper J N. Identification of a novel family of cell-surface proteins expressed in human vascular endothelium. J Biol Chem. 2002 Nov. 29; 277(48):46364-73. Epub 2002 Sep. 21. PMID: 12270931

  • Grimmond S, Larder R, Van Hateren N, Siggers P, Hulsebos T J, Arkell R, Greenfield A. Cloning, mapping, and expression analysis of a gene encoding a novel mammalian EGF-related protein (SCUBE1). Genomics. 2000 Nov. 15; 70(1):74-81. PMID: 11087664

  • Stadler F, Kolb G, Rubusch L, Baker S P, Jones E G, Akbarian S. Histone methylation at gene promoters is associated with developmental regulation and region-specific expression of ionotropic and metabotropic glutamate receptors in human brain. J Neurochem. 2005 July; 94(2):324-36. PMID: 15998284

  • Bah J, Quach H, Ebstein R P, Segman R H, Melke J, Jamain S, Rietschel M, Modai I, Kanas K, Karni O, Lerer B, Gourion D, Krebs M O, Etain B, Schurhoff F, Szoke A, Leboyer M, Bourgeron T. Maternal transmission disequilibrium of the glutamate receptor GRIK2 in schizophrenia. Neuroreport. 2004 Aug. 26; 15(12):1987-91. PMID: 15305151

  • Sinclair P B, Sorour A, Martineau M, Harrison C J, Mitchell W A, O'Neill E, Foroni L. A fluorescence in situ hybridization map of 6q deletions in acute lymphocytic leukemia: identification and analysis of a candidate tumor suppressor gene. Cancer Res. 2004 Jun. 15; 64(12):4089-98. PMID: 15205317

  • Wolf S, Janzen A, Vekony N, Martine U, Strand D, Closs E I. Expression of solute carrier 7A4 (SLC7A4) in the plasma membrane is not sufficient to mediate amino acid transport activity. Biochem J. 2002 Jun. 15; 364(Pt 3):767-75. PMID: 12049641

  • Sperandeo M P, Borsani G, Incerti B, Zollo M, Rossi E, Zuffardi O, Castaldo P, Taglialatela M, Andria G, Sebastio G. The gene encoding a cationic amino acid transporter (SLC7A4) maps to the region deleted in the velocardiofacial syndrome. Genomics. 1998 Apr. 15; 49(2):230-6. PMID: 9598310

  • Peyrl A, Krapfenbauer K, Slavc I, Strobel T, Lubec G. Proteomic characterization of the human cortical neuronal cell line HCN-2. J Chem Neuroanat. 2003 November; 26(3):171-8. PMID: 14615026

  • Niki T, Takahashi-Niki K, Taira T, Iguchi-Ariga S M, Ariga H. DJBP: a novel DJ-1-binding protein, negatively regulates the androgen receptor by recruiting histone deacetylase complex, and DJ-1 antagonizes this inhibition by abrogation of this complex. Mol Cancer Res. 2003 February; 1(4):247-61. PMID: 12612053

  • Killebrew D A, Troelstrup D, Shiramizu B. Preferential HIV-1 integration sites in macrophages and HIV-associated malignancies. Cell Mol Biol (Noisy-1e-grand). 2004; 50 Online Pub:OL581-9. PMID: 15555424

  • Shou S, Scott V, Reed C, Hitzemann R, Stadler H S. Transcriptome analysis of the murine forelimb and hindlimb autopod. Dev Dyn. 2005 September; 234(1):74-89. PMID: 16059910 [PubMed—in process]

  • Plageman T F Jr, Yutzey K E. T-box genes and heart development: putting the “T” in heart. Dev Dyn. 2005 January; 232(1):11-20. Review. PMID: 15580613

  • Bussen M, Petry M, Schuster-Gossler K, Leitges M, Gossler A, Kispert A. The T-box transcription factor Tbx18 maintains the separation of anterior and posterior somite compartments. Genes Dev. 2004 May 15; 18(10):1209-21. PMID: 15155583

  • Rosemann M, Kuosaite V, Nathrath M, Strom T M, Quintanilla-Martinez L, Richter T, Imai K, Atkinson M J. Allelic imbalance at intragenic markers of Tbx18 is a hallmark of murine osteosarcoma. Carcinogenesis. 2003 March; 24(3):371-6. PMID: 12663494

  • Ding L H, Shingyoji M, Chen F, Hwang J J, Burma S, Lee C, Cheng J F, Chen D J. Gene expression profiles of normal human fibroblasts after exposure to ionizing radiation: a comparative study of low and high doses. Radiat Res. 2005 July; 164(1):17-26. PMID: 15966761

  • Yankee T M, Draves K E, Clark E A. Expression and function of the adaptor protein Gads in murine B cells. Eur J Immunol. 2005 April; 35(4):1184-92. PMID: 15761845

  • Shiraiwa H, Takei M, Yoshikawa T, Azuma T, Kato M, Mitamura K, Ueki T, Kida A, Horie T, Seki N, Sawada S. Detection of Grb-2-related adaptor protein gene (GRAP) and peptide molecule in salivary glands of MRL/lpr mice and patients with Sjogren's syndrome. J Int Med Res. 2004 May-June; 32(3):284-91. PMID: 15174222

  • Guyot B, Mouchiroud G. Characterization of promoter elements directing Mona/Gads molecular adapter expression in T and myelomonocytic cells: involvement of the AML-1 transcription factor. J Leukoc Biol. 2003 February; 73(2):263-72. PMID: 12554803

  • Guyot B, Arnaud S, Phothirath P, Bourette R P, Grasset M F, Rigal D, Mouchiroud G. Genomic organization and restricted expression of the human Mona/Gads gene suggests regulation by two specific promoters. Gene. 2002 May 15; 290(1-2):173-9. PMID: 12062812

  • Chou H H, Hayakawa T, Diaz S, Krings M, Indriati E, Leakey M, Paabo S, Satta Y, Takahata N, Varki A. Inactivation of CMP-N-acetylneuraminic acid hydroxylase occurred prior to brain expansion during human evolution. Proc Natl Acad Sci USA. 2002 Sep. 3; 99(18):11736-41. Epub 2002 Aug. 21. PMID: 12192086

  • Igaz P, Salvi R, Rey J P, Glauser M, Pralong F P, Gaillard R C. Effects of cytokines on GnRH gene expression in primary hypothalamic neurons and in GnRH neurons immortalized conditionally. Endocrinology. 2005 Nov. 10; [Epub ahead of print] PMID: 16282355 [PubMed—as supplied by publisher]

  • Loewen G M, Tracy E, Blanchard F, Tan D, Yu J, Raza S, Matsui S, Baumann H. Transformation of human bronchial epithelial cells alters responsiveness to inflammatory cytokines. BMC Cancer. 2005 Nov. 4; 145. PMID: 16271139 [PubMed—in process]

  • Znoyko I, Sohara N, Spicer S S, Trojanowska M, Reuben A. Expression of oncostatin M and its receptors in normal and cirrhotic human liver. J Hepatol. 2005 November; 43(5):893-900. Epub 2005 Jul. 26. PMID: 16169119 [PubMed—in process]

  • Finelt N, Gazel A, Gorelick S, Blumenberg M. Transcriptional responses of human epidermal keratinocytes to Oncostatin-M. Cytokine. 2005 Aug. 21; 31(4):305-13. PMID: 16023359

  • Kong W, Abidi P, Kraemer F B, Jiang J D, Liu J. In vivo activities of cytokine oncostatin M in the regulation of plasma lipid levels. J Lipid Res. 2005 June; 46(6):1163-71. Epub 2005 Mar. 16. PMID: 15772430

  • Znoyko I, Sohara N, Spicer S S, Trojanowska M, Reuben A. Comparative studies of oncostatin M expression in the tissues of adult rodents. Anat Rec A Discov Mol Cell Evol Biol. 2005 March; 283(1):182-6. PMID: 15674824

  • Li C, Zhang F, Lin M, Liu J. Induction of S100A9 gene expression by cytokine oncostatin M in breast cancer cells through the STAT3 signaling cascade. Breast Cancer Res Treat. 2004 September; 87(2):123-34. PMID: 15377837

  • Holzer R G, Ryan R E, Tommack M, Schlekeway E, Jorcyk C L. Oncostatin M stimulates the detachment of a reservoir of invasive mammary carcinoma cells: role of cyclooxygenase-2. Clin Exp Metastasis. 2004; 21(2):167-76. PMID: 15168734

  • Cross A, Edwards S W, Bucknall R C, Moots R J. Secretion of oncostatin M by neutrophils in rheumatoid arthritis. Arthritis Rheum. 2004 May; 50(5):1430-6. PMID: 15146412

  • Gwechenberger M, Moertl D, Pacher R, Huelsmann M. Oncostatin-M in myocardial ischemia/reperfusion injury may regulate tissue repair. Croat Med J. 2004 April; 45(2):149-57. PMID: 15103750

  • Levy M T, Trojanowska M, Reuben A. Oncostatin M: a cytokine upregulated in human cirrhosis, increases collagen production by human hepatic stellate cells. J Hepatol. 2000 February; 32(2):218-26. PMID: 10707861

  • Vasse M, Pourtau J, Trochon V, Muraine M, Vannier J P, Lu H, Soria J, Soria C. Oncostatin M induces angiogenesis in vitro and in vivo. Arterioscler Thromb Vasc Biol. 1999 August; 19(8):1835-42. PMID: 10446061

  • Battini R, Battaglia A, Bertini V, Cioni G, Parrini B, Rapalini E, Simi P, Tinelli F, Valetto A. Characterization of the phenotype and definition of the deletion in a new patient with ring chromosome 22. Am J Med Genet A. 2004 Oct. 1; 130(2):196-9. PMID: 15372517

  • Formstone C J, Mason I. Expression of the Celsr/flamingo homologue, c-fmi1, in the early avian embryo indicates a conserved role in neural tube closure and additional roles in asymmetry and somitogenesis. Dev Dyn. 2005 February; 232(2):408-13. PMID: 15614764

  • Tissir F, De-Backer O, Goffinet A M, Lambert de Rouvroit C. Developmental expression profiles of Celsr (Flamingo) genes in the mouse. Mech Dev. 2002 March; 112(1-2):157-60. PMID: 11850187

  • Bacon L, Eagle R A, Meyer M, Easom N, Young N T, Trowsdale J. Two human ULBP/RAET1 molecules with transmembrane regions are ligands for NKG2D. J Immunol. 2004 Jul. 15; 173(2):1078-84. PMID: 15240696

  • Jan Chalupny N, Sutherland C L, Lawrence W A, Rein-Weston A, Cosman D. ULBP4 is a novel ligand for human NKG2D. Biochem Biophys Res Commun. 2003 May 23; 305(1):129-35. PMID: 12732206

  • Eckfeld K, Hesson L, Vos M D, Bieche I, Latif F, Clark G J. RASSF4/AD037 is a potential ras effector/tumor suppressor of the RASSF family. Cancer Res. 2004 Dec. 1; 64(23):8688-93. PMID: 15574778

  • Chow L S, Lo K W, Kwong J, Wong A Y, Huang D P. Aberrant methylation of RASSF4/AD037 in nasopharyngeal carcinoma. Oncol Rep. 2004 October; 12(4):781-7. PMID: 15375500

  • Futscher B W, Oshiro M M, Wozniak R J, Holtan N, Hanigan C L, Duan H, Domann F E. (2002) Role for DNA methylation in the control of cell type specific maspin expression. Nat Genet. 31(2):175-9.


Claims
  • 1. A method for classifying a biological sample, comprising: obtaining a biological sample from a subject;determining the expression status of at least one gene or genomic sequence selected from the group consisting the genes or genomic sequences according to Table 1 in said sample; andclassifying said biological sample according to said expression status.
  • 2. The method according to claim 1, wherein the class is selected from the group consisting of organ type, tissue type, cell type and disease state.
  • 3. The method according to claim 2, wherein said class consists of T-lymphocytes.
  • 4. The method according to claim 3, wherein said genomic sequences are selected from at least one of Tables 8A and 9A.
  • 5. The method according to claim 2, wherein said class consists of embryonic liver.
  • 6. The method according to claim 5, wherein said genomic sequences are selected from at least one of Tables 8B and 9B.
  • 7. The method according to claim 2, wherein said class consists of embryonic skeletal muscle.
  • 8. The method according to claim 7, wherein said genomic sequences are selected from at least one of Tables 8C and 9C.
  • 9. The method according to claim 2, wherein said class consists of fibroblasts.
  • 10. The method according to claim 9, wherein said genomic sequences are selected from at least one of Tables 8D and 9D.
  • 11. The method according to claim 2, wherein said class consists of heart muscle.
  • 12. The method according to claim 11, wherein said genomic sequences are selected from at least one of Tables 8E and 9E.
  • 13. The method according to claim 2, wherein said classes are heart muscle and skeletal muscle.
  • 14. The method according to claim 13, wherein said genomic sequences are selected from Table 8F.
  • 15. The method according to claim 2, wherein said class consists of keratinocytes.
  • 16. The method according to claim 15, wherein said genomic sequences are selected from at least one of Tables 8G and 9F.
  • 17. The method according to claim 2, wherein said class consists of liver.
  • 18. The method according to claim 17, wherein said genomic sequences are selected from at least one of Tables 8H and 9G.
  • 19. The method according to claim 2, wherein said class consists of melanocytes.
  • 20. The method according to claim 19, wherein said genomic sequences are selected from at least one of Table 8I and 9H.
  • 21. The method according to claim 2, wherein said class consists of placenta.
  • 22. The method according to claim 21, wherein said genomic sequences are selected from at least one of Table 8J and 9I.
  • 23. The method according to claim 2, wherein said class consists of skeletal muscle.
  • 24. The method according to claim 23, wherein said genomic sequences are selected from at least one of Table 8K and 9J.
  • 25. The method according to claim 2, wherein said class consists of sperm.
  • 26. The method according to claim 25, wherein said genomic sequences are selected from Table 8L.
  • 27. The method according to claim 1, wherein the sample is selected from the group consisting of cells or cell lines, histological slides, biopsies, paraffin-embedded tissue, bodily fluids, sputum, stool, nipple aspirate, cerebrospinal fluid, ejaculate, urine, blood, and combinations thereof.
  • 28. The method according to claim 1, wherein the expression is determined by measuring the level of at least one of mRNA, cDNA or polypeptide.
  • 29. The method according to claim 1, wherein the expression is determined by use of at least one technique selected from the group of Northern blot analysis, reverse transcriptase PCR, real-time PCR, RNAse protection, and microarray.
  • 30. The method according to claim 1, wherein said expression is determined by determining the level of methylation or methylation status of one or more CpG positions within said genes or genomic regions.
  • 31. The method according to claim 30, comprising contacting genomic DNA isolated from a biological sample, with at least one reagent, or series of reagents that distinguishes between methylated and non-methylated CpG dinucleotides within at least one target region of the genomic DNA, wherein the target region comprises, or hybridizes under stringent conditions to a sequence of at least 16 contiguous nucleotides of at least one gene or genomic sequence selected from Table 1, wherein said contiguous nucleotides comprise at least one CpG dinucleotide sequence, and wherein classification of said sample is, at least in part, afforded.
  • 32. The method according to claim 31, comprising: isolating genomic DNA from a biological sample taken from a subject;treating the genomic DNA, or a portion or fragment thereof, with one or more reagents to convert 5-position unmethylated cytosine bases to uracil or to another base that is detectably dissimilar to cytosine in terms of hybridization properties;contacting the treated genomic DNA, or the treated portion or fragment thereof, with an amplification enzyme and at least two primers comprising, in each case a contiguous sequence at least 18 nucleotides in length that is complementary to, or hybridizes under moderately stringent or stringent conditions to a converted sequence selected from Table 1 and complements thereof, wherein the treated DNA or a fragment thereof is either amplified to produce one or more amplificates, or is not amplified;determining, based on the presence or absence of, or on the quantity or on a property of said amplificate, the methylation state of at least one CpG dinucleotide sequence of at least one gene or sequence selected from Table 1, or an average, or a value reflecting an average methylation state of a plurality of CpG dinucleotide sequences of at least one gene or sequence selected from Table 1; andclassifying said sample according to said methylation state.
  • 33. A treated nucleic acid derived from the genomic sequences of Table 2, wherein the treatment is suitable to convert at least one unmethylated cytosine base of the genomic DNA sequence to uracil or another base that is detectably dissimilar to cytosine in terms of hybridization.
  • 34. A nucleic acid, comprising at least 16 contiguous nucleotides of a treated genomic DNA sequence selected from the converted sequences according to Table 1 and sequences complementary thereto, wherein said nucleic acid is not identical or complementary to a genomic sequence according to Table 2, and wherein the treatment is suitable to convert at least one unmethylated cytosine base of the genomic DNA sequence to uracil or another base that is detectably dissimilar to cytosine in terms of hybridization.
  • 35. The nucleic acid of any one of claims 33 and 34, wherein the contiguous base sequence comprises at least one CpG, TpG or CpA dinucleotide sequence.
  • 36. The nucleic acid of claim 35, wherein the treatment comprises use of a reagent selected from the group consisting of bisulfite, hydrogen sulfite, disulfite, and combinations thereof.
  • 37. An oligomer, comprising a sequence of at least 9 contiguous nucleotides that is complementary to, or hybridizes under moderately stringent or stringent conditions to a treated genomic DNA sequence selected from the converted sequences according to Table 2 and sequences complementary thereto, wherein said nucleic acid is not identical or complementary to a genomic sequence according to Table 2.
  • 38. The oligomer of claim 37, comprising at least one CpG, CpA or TpG dinucleotide sequence.
  • 39. A kit for use in for use in the classification of a biological sample, the kit comprising means for detecting the polypeptides of a gene or genomic region selected from Table 1.
  • 40. The kit according to claim 39, further comprising: a container suitable for containing the means for detecting polypeptides, and a biological sample of the patient comprising said polypeptides, wherein the means for detecting polypeptides can from complexes with the polypeptides; anda means to detect the complexes.
  • 41. A kit for use in the classification of a biological sample, comprising means for measuring the level of mRNA transcription of a gene or genomic region selected from Table 1.
  • 42. The kit according to claim 41, further comprising: a container suitable for containing the means for measuring the level of mRNA, and a biological sample of the patient comprising mRNA of a gene or genomic region selected from Table 1 wherein the means for measuring the level of mRNA are able to hybridize to the mRNA; anda means for detecting the hybridized mRNA complexes.
  • 43. A kit comprising: a) at least one bisulfite reagent selected from the group consisting of bisulfite hydrogen sulfite, and disulfite; andb) at least two nucleic acid molecules comprising, in each case a contiguous sequence at least 16 nucleotides that is complementary to, or hybridizes under moderately stringent or stringent conditions to a converted sequence selected from Table 2, and complements thereof.
  • 44. A composition comprising the following: a nucleic acid comprising a sequence at least 18 bases in length of a segment of the converted genomic DNA according to one of the converted sequences of Table 2 and sequences complementary thereto; anda buffer comprising at least one of magnesium chloride, dNTP, taq polymerase, an oligomer, in particular an oligonucleotide or peptide nucleic acid (PNA)-oligomer, said oligomer comprising in each case at least one base sequence having a length of at least 9 nucleotides which is complementary to, or hybridizes under moderately stringent or stringent conditions to a converted genomic DNA according to one of the converted sequences of Table 1 and sequences complementary thereto.
  • 45. (canceled)
  • 46. The kit of claim 40, further comprising instructions for use and interpretation of the kit results.
  • 47. The kit of claim 42, further comprising instructions for use and interpretation of the kit results.
Priority Claims (3)
Number Date Country Kind
050213313.3 Sep 2005 EP regional
05090346.7 Dec 2005 EP regional
06090110.5 Jun 2006 EP regional
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/EP2006/009454 9/28/2006 WO 00 7/2/2008