COMPOSITIONS AND METHODS FOR DIAGNOSIS OR PROGNOSIS OF TESTICULAR CANCER

Information

  • Patent Application
  • 20100124747
  • Publication Number
    20100124747
  • Date Filed
    November 03, 2009
    14 years ago
  • Date Published
    May 20, 2010
    14 years ago
Abstract
Provided are compositions and methods for diagnosis or prognosis of testicular or male germ-cell derived cancer, comprising: obtaining sperm DNA from a test subject; determining the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and thereby determining or diagnosing testicular or male germ-cell derived cancer. Provided are compositions and methods for identifying agents that cause testicular or male germ-cell derived cancer, comprising: obtaining human ES-cell derived primordial germ cells; contacting the germ cells or descendants thereof, with a test agent; culturing the contacted cells; determining, using a genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the above group; and identifying at least one test agent that causes testicular or male germ-cell derived cancer.
Description
FIELD OF THE INVENTION

Particular aspects relate generally to DNA methylation and epigenetic reprogramming during development, gametogenesis and cancer, and more particularly to novel and effective epigenetic biomarkers and methods for diagnosis or prognosis of testicular or male germ-cell derived cancer, comprising determining the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1. Additional aspects relate to compositions and methods for identifying and/or screening for agents that cause testicular or male germ-cell derived cancer, comprising contacting human (or murine, rat, Etc.) ES-cell derived primordial germ cells with a test agent and determining the methylation status of at least one CpG dinucleotide sequence from at least one sequence as disclosed herein.


BACKGROUND

Ten to twenty percent of couples attempting pregnancy are infertile. Male-factor infertility accounts entirely for approximately 20% of these cases, and is contributory in an additional 30% [1,2]. Well defined causes of male-factor infertility are known to include congenital and acquired dysfunction of the hypothalamic-pituitary-testicular endocrine axis, anatomic defects, chromosomal abnormalities, and point mutations [3-5]. However, these diagnoses account for only a small proportion of cases, and etiology remains unknown for most male-factor infertility patients [1,2].


The mammalian germ line undergoes extensive epigenetic reprogramming during development and gametogenesis. In males, dramatic chromatin remodeling occurs during spermatogenesis [6,7], and widespread erasure of DNA methylation followed by de novo DNA methylation occurs developmentally in two broad waves [6, 8-11]. The first occurs before emergence of the germ line, establishing a pattern of somatic-like DNA hypermethylation in cells of the pre-implantation embryo that are destined to give rise to all cells of the body, including germ cells. The second widespread occurrence of erasure takes place uniquely in primordial germ cells. Subsequent de novo methylation occurs during germ cell maturation and spermatogenesis, establishing a male germ line pattern of DNA methylation that remains hypomethylated compared with somatic cell DNA [8, 12-16].


A small number of studies have addressed the epigenetic state of the human male germ line. Substantial variation in DNA methylation profiles is reported in ejaculated sperm of young, apparently healthy men. Notable distinctions were observed both between samples from separate men and among individually assayed sperm from the same man [17].


Although this variation suggests that DNA methylation may be used as a biomarker of sperm quality, semen quality and fertility were not assessed in this study [17].


SUMMARY OF EXEMPLARY ASPECTS

Male-factor infertility is a common condition, and etiology is unknown for a high proportion of cases. Abnormal epigenetic programming of the germline is disclosed as a mechanism compromising spermatogenesis of some men currently diagnosed with idiopathic infertility. During germ cell maturation and gametogenesis, cells of the germ line undergo extensive epigenetic reprogramming. This process involves widespread erasure of somatic-like patterns of DNA methylation followed by establishment of sex-specific patterns by de novo DNA methylation.


According to particular aspects, incomplete reprogramming of the male germ line results in both altered sperm DNA methylation and compromised spermatogenesis.


Particular aspects provide the first discovery and disclosure ever of a broad epigenetic defect associated with abnormal semen parameters. Additional aspects relate to an underlying mechanism for these broad epigenetic changes, comprising improper erasure of DNA methylation during epigenetic reprogramming of the male germ line.


Concentration, motility and morphology of sperm was determined in semen samples collected by male members of couples attending an infertility clinic. METHYLIGHT™ and ILLUMINA™ assays were used to measure methylation of DNA isolated from purified sperm from the same samples. Methylation at numerous sequences was elevated in DNA from poor quality sperm, and provide novel and effective epigenetic biomarkers of sperm quality, semen quality and fertility.


Particular exemplary aspects, provide methods for determining or diagnosing abnormal sperm or fertility, comprising: obtaining a sample of human sperm DNA from a test subject; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and determining, based on the methylation status of the at least one CpG sequence, the presence or diagnosis of abnormal sperm or fertility with respect to the test subject. In certain aspects, the determined methylation status of the at least one CpG sequence is hypermethylation. In particular embodiments, determining the methylation status of at least one CpG dinucleotide sequence comprises 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. Preferably, treating comprises use of bisulfite treatment of the DNA.


In certain aspects, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


In particular aspects, abnormal sperm comprises at least one of abnormal sperm concentration, abnormal motility, abnormal total normal morphology, abnormal volume, and abnormal viscosity. In certain embodiments, abnormal sperm comprises at least one of abnormal sperm concentration, abnormal motility, and abnormal total normal morphology.


Certain aspects of the methods, comprise determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8 and PLAGL1. In certain embodiments, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, and PLAGL1 SEQ ID NOS:7 and 19.


Yet additional aspects, provide methods for determining or diagnosing abnormal sperm or fertility, comprising: obtaining a sample of human sperm DNA from a test subject; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence from each of a repetitive DNA element sequence group, a maternally imprinted gene sequence group, and a non-imprinted gene sequence group; and determining, based on the methylation status of the at least one CpG sequence from each of the groups, the presence or diagnosis of abnormal sperm or fertility with respect to the test subject. In certain implementations, the at least one gene sequence from a repetitive element group comprises at least one selected from the group consisting of SAT2CHRM1 SEQ ID NOS:9 and 21. In certain aspects, the at least one gene sequence from a maternally imprinted gene group comprises at least one selected from the group consisting of PLAGL1 SEQ ID NOS:7 and 19, MEST SEQ ID NOS:5 and 17, and DIRAS3 SEQ ID NOS:3 and 15. In particular embodiments, the at least one gene sequence from a non-imprinted gene group comprises at least one selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, SFN SEQ ID NOS:6 and 18, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


Yet further aspects provide methods for screening for agents that cause spermatogenic deficits, abnormal sperm or abnormal fertility comprising: obtaining human ES-cell derived primordial germ cells; contacting the germ cells or descendants thereof, with at least one test agent; culturing the contacted germ cells or the descendants thereof under conditions suitable for germ cell proliferation or development; obtaining a sample of genomic DNA from the contacted cultured germ cells or the descendants thereof; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and identifying, based on the methylation status of the at least one CpG sequence, at least one test agent that causes at least one of spermatogenic deficits, abnormal sperm, and abnormal fertility. In certain aspects, the determined methylation status of the at least one CpG sequence is hypermethylation. In certain embodiments, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


Additional aspects provide methods for diagnosis or prognosis of testicular or male germ-cell derived cancer, comprising: obtaining a sample of human sperm DNA from a test subject; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and determining, based on the methylation status of the at least one CpG sequence, diagnosis or prognosis of testicular or male germ-cell derived cancer with respect to the test subject. In certain aspects, the determined methylation status of the at least one CpG sequence is hypermethylation. In particular aspects, determining the methylation status of at least one CpG dinucleotide sequence comprises 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. In certain embodiments, treating comprises use of bisulfite treatment of the DNA. In particular aspects, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


In certain aspects, the diagnosis or prognosis is of germ-cell derived testicular cancer.


Particular embodiments of the cancer diagnostic or prognostic assays comprise determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8 and PLAGL1. In certain aspects, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, and PLAGL1 SEQ ID NOS:7 and 19.'


Additional aspects provide a method for diagnosis or prognosis of testicular or male germ-cell derived cancer, comprising: obtaining a sample of human sperm DNA from a test subject; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence from each of a repetitive DNA element sequence group, a maternally imprinted gene sequence group, and a non-imprinted gene sequence group; and determining, based on the methylation status of the at least one CpG sequence from each of the groups, diagnosis or prognosis of testicular or male germ-cell derived cancer with respect to the test subject. In certain aspects, the determined methylation status of the at least one CpG sequence is hypermethylation. In particular implementations, the methylation status of at least one CpG dinucleotide sequence comprises 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. In certain embodiments, treating comprises use of bisulfite treatment of the DNA. In certain aspects, the at least one gene sequence from a repetitive element group comprises at least one selected from the group consisting of SAT2CHRM1 SEQ ID NOS:9 and 21. In particular aspects, the at least one gene sequence from a maternally imprinted gene group comprises at least one selected from the group consisting of PLAGL1 SEQ ID NOS:7 and 19, MEST SEQ ID NOS:5 and 17, and DIRAS3 SEQ ID NOS:3 and 15. In certain embodiments, the at least one gene sequence from a non-imprinted gene group comprises at least one selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, SFN SEQ ID NOS:6 and 18, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


Additionally provided are methods for screening for agents that cause testicular or male germ-cell derived cancer, comprising: obtaining human ES-cell derived primordial germ cells; contacting the germ cells or descendants thereof, with at least one test agent; culturing the contacted germ cells or the descendants thereof under conditions suitable for germ cell proliferation or development; obtaining a sample of genomic DNA from the contacted cultured germ cells or the descendants thereof; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and identifying, based on the methylation status of the at least one CpG sequence, at least one test agent that causes testicular or male germ-cell derived cancer. In certain aspects, the determined methylation status of the at least one CpG sequence is hypermethylation. In certain aspectgs, determining the methylation status of at least one CpG dinucleotide sequence comprises 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. In certain embodiments, treating comprises use of bisulfite treatment of the DNA. In particular embodiments, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24. In certain aspects, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, and PLAGL1 SEQ ID NOS:7 and 19.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows, according to particular exemplary aspects, box plots illustrating associations between semen parameters and level of methylation (PMR) in DNA isolated from 65 study sperm samples. DNA methylation was measured by MethyLight. Methylation targets were sequences specific to the genes HRAS, NTF3, MT1A, PAX8, PLAGL1, DIRAS3, MEST and SFN and the repetitive element Satellite 2 (SAT2CHRM1). P-value for trend over category of semen parameter is given for each plot. Rows: DNA methylation targets; columns: semen parameters.



FIG. 2 shows, according to particular exemplary aspects, cluster analysis of 36 MethyLight targets in 65 study sperm DNA samples. Left: dendrogram defining clusters; rows: 35 methylation targets; columns: 65 study samples ordered left to right on sperm concentration (samples A-G were also included in Illumina analyses (see FIG. 3)) with poor to good concentration (blue), motility (purple), and morphology (green) represented by darkest to lightest hue; body of figure: standardized PMR values represented lowest to highest as yellow to red. X=missing.



FIG. 3 shows, according to particular exemplary aspects, Results of Illumina analysis of 1,421 autosomal sequences in DNA isolated from sperm and buffy coat. Seven study sperm samples (A-G; ordered left to right on sperm concentration), screening sperm (S), two buffy coat (1-2). Level of DNA methylation scored as β-value. Color: β-value for column sample at row sequence (green: β<0.1; yellow: 0.1≦β≦0.25; orange 0.25<β≦30.5; red: β>0.5). MI and PI: maternally and paternally imprinted genes (black bar). Sequences assigned to tertile of median β-value among buffy coat DNA samples (I, II, III) and sorted within tertile on median β-value among sperm DNA samples. Box 1: sequences with sperm-specific DNA methylation; Box 2: sequences with buffy coat-specific DNA methylation.



FIGS. 4
a, b and c schematically depict mammalian primordial germ line development, showing that primordial germ cells undergo extensive epigenetic reprogramming during development, where widespread erasure of DNA methylation occurs in primordial germ cells as they migrate to the gonadal ridges). After the testes form, de novo DNA methylation establishes male-specific DNA methylation during germ cell maturation and spermatogenesis (see FIGS. 4a and b). According to particular aspects of the present invention, DNA methylation of sperm from men with poor fertility is consistent with disruption of the widespread erasure of DNA methylation that occurs early in the development of germ cells (see FIG. 4c).





DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS

Overview. There have been several prior art attempts in the art to assess sperm DNA methylation together with either sperm quality or fertility outcomes. However, the measures of DNA methylation used were limited, consisting of either a nonspecific genome-wide measure [18], or small and specialized subsets of DNA methylation targets [19-21].


Specifically, in the only study prior art study addressing the relationship between DNA methylation and fertility outcomes, immunostaining was used to measure genome-wide levels of DNA methylation in samples of ejaculated sperm collected for conventional in vitro fertilization (IVF) [18], and no association was observed between sperm DNA methylation and either fertilization rate or embryo quality in 63 IVF cycles. There was, however, a possible association with pregnancy rate after transfer of good quality embryos. Interpretation of these results is limited by both small sample size and the use of a single summary measure of genome-wide DNA methylation.


Moreover, with respect to the prior art studies [19-21] with small and specialized subsets of DNA methylation targets, sequence-specific measures were used to investigate the relationship between methylation of human sperm DNA and spermatogenesis. One study assessed DNA from spermatogonia and spermatocytes microdissected from seminiferous tubules of biopsied testicular tissue with spermatogenic arrest. DNA profiles consistent with correctly established paternal imprints were reported in all samples [19]. In the remaining two studies [20 and 21], DNA profiles were measured at specific DMRs associated with each of two genes, one paternally and one materially imprinted, and the resulting profiles were related to concentration of ejaculated sperm, an indicator of sperm quality. One of these studies reported correctly erased maternal imprints and correctly established paternal imprints in DNA from sperm of low concentration [21]. By contrast, the second reported that although maternal imprinting of MEST was correctly erased in DNA from sperm of low concentration, methylation at an H19 sequence typically de novo methylated in spermatogenesis was incomplete in these samples [20]. No compelling explanation was offered for the apparently differing results of these studies. It is noteworthy, however, that each addressed sequences of only one or two imprinted genes, an extremely small and specialized subset of DNA methylation targets in the human genome. Data from these published studies could not, therefore, have revealed a disruption involving large numbers of genes, or shown that genes that are not imprinted are also affected.


Particular aspects provide methods for determining or diagnosing abnormal sperm or fertility, comprising: obtaining a sample of human sperm DNA from a test subject; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and determining, based on the methylation status of the at least one CpG sequence, the presence or diagnosis of abnormal sperm or fertility with respect to the test subject. In certain embodiments the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


In particular aspects at least on CpG dinucleotide sequence within an amplicon is determined. In preferred aspects, the at least one amplicon sequence is selected from the group consisting of: HRAS SEQ ID NOS:20, NTF3 SEQ ID NO: 14, MT1A SEQ ID NO:16, PAX8 SEQ ID NO:13, DIRAS3 SEQ ID NO:15, PLAGL1 SEQ ID NO:19, SFN SEQ ID NO:18, SAT2CHRM1 SEQ ID NO:21, MEST SEQ ID NO:17, RNR1 SEQ ID NO:22, CYP27B1 SEQ ID NO:23 and ICAM1 SEQ ID NO:24.


Preferably, the amplicon is part of a contiguous CpG island sequence. In preferred aspects, the CpG island sequence sequence is selected from the group consisting of: HRAS SEQ ID NOS:63, NTF3 SEQ ID NO:2, MT1A SEQ ID NO:4, PAX8 SEQ ID NO:1, DIRAS3 SEQ ID NO:3, PLAGL1 SEQ ID NO:7, SFN SEQ ID NO:6, SAT2CHRM1 SEQ ID NO:9, MEST SEQ ID NO:5, RNR1 SEQ ID NO:10, CYP27B1 SEQ ID NO:11 and ICAM1 SEQ ID NO:12.


Coordinate methylation within CpG islands. According to particular aspects, and as recognized in the relevant art, hypermethylation is coordinate within a CpG island. For Example, data (see Eckhardt et al., Nat Genet. 2006 December; 38(12):1378-85. Epub 2006 Oct. 29; incorporated by reference herein in its entirety) has been generated by analyzing methylation (using bisulfite sequencing) in CG-rich regions across entire chromosomes to provide a methylation map of the human genome (at least of the CPG rich regions thereof). To date, these data comprise methylation data of 3 complete human chromosomes (22, 20, and 6) for a variety of different tissues and cell types. Based on these data, for methylation patterns within CpG dense regions, methylation is typically found to be either present for all methylatable cytosines or none. This methylation characteristic or pattern is referred to in the art as “co-methylation” or “coordinate methylation.” The findings of this paper support a “significant correlation” of comethylation over the distance of at least 1,000 nucleotides in each direction from a particular determined CpG within a CpG dense region (see, e.g., page 2, column 2, 1st full paragraph, of Eckhardt et al publication document). Furthermore, such co-methylation forms the basis for long-standing common methods such as MSP and particular MethyLight embodiments that rely on such co-methylation (e.g., as employed herein, the primers and/or probes each typically encompass multiple CpG sequences), and has now been further confirmed over entire chromosomes by Eckhardt et al. Therefore, in view of the teachings of the present specification, there is a reasonable correlation between the claimed coordinately methylated sequences, and the recited methods and exemplary methylation marker sequences.


Measurement of DNA Methylation of the Genomic DNA of Spermatozoa at CpG Islands, DMRs of Imprinted Genes and Repetitive Elements

The present specification describes and discloses the first study ever to investigate the epigenetic state of abnormal human sperm using an extensive panel of DNA methylation assays. Abnormal epigenetic programming of the germ line is herein disclosed as a mechanism compromising fertility of particular men currently diagnosed with idiopathic infertility. Aspects of the present invention indicate that one or more epigenetic processes lead to abnormal spermatogenesis and compromised sperm function.


To assess sperm DNA, methylation at specific targets that are both more numerous and less specialized, a relatively large set of sequence-specific assays was selected for use in the presently disclosed studies and invention.


Specifically, DNA methylation was measured in ejaculated spermatozoa-interrogating sequences in repetitive elements, promoter CpG islands, and differentially methylated regions (DMRs) of imprinted genes. Then, to address the possible role of epigenetic programming in abnormal human spermatogenesis, sequence-specific levels of DNA methylation were related to standard measures of sperm quality.


Applicants' observations indicate a broad epigenetic abnormality of poor quality human sperm in which levels of DNA methylation are elevated at numerous sequences in several genomic contexts. Previous studies of DNA methylation in poor quality sperm interrogated only imprinted loci, measuring methylation of sequences in only one or two genes [19-21].


Aspects of the present invention provide, inter alia, compositions and methods having substantial utility for diagnosing or determining the presence of abnormal sperm or fertility (e.g., comprising at least one of abnormal sperm concentration, abnormal total normal morphology, abnormal motility, abnormal volume, and abnormal viscosity).


As described in the working Example 1, herein below, Applicants initially evaluated 294 MethyLight reactions for the presence of methylation in sperm DNA from an anonymous semen sample obtained from a sperm bank. Standard semen analysis was then conducted on samples collected by 69 men during clinical evaluation of couples with infertility. Thirty seven selected MethyLight reactions were used to assay sperm DNA from 65 of the study samples.


At many of the 37 sequences, methylation levels were elevated in DNA from poor quality sperm. For example, striking associations with each of sperm concentration, motility and morphology were observed for five sequences: HRAS, NTF3, MT1A, PAX8 and the maternally imprinted gene PLAGL1 (FIG. 1). Applicants also found elevated DNA methylation to be significantly associated with poor semen parameters for the DIRAS3 and MEST maternally imprinted genes (FIG. 1).


Associations between results of each of the 37 MethyLight assays and sperm concentration were highly significant for HRAS, NTF3, MT1A, PAX8, DIRAS3 and PLAGL1 and were also significant (somewhat less) for SFN, SAT2CHRM1 and MEST (see Table 1 of Example 1, and see also FIG. 1).


Unsupervised cluster analysis identified three distinct clusters of sequences based on DNA methylation profiles in the 65 samples (FIG. 2). The middle cluster shown in FIG. 2 includes eight of the above nine sequences (all except MT1A) individually associated with semen parameters, and includes not only three sequences that are differentially methylated on imprinted loci, but also three single copy sequences specific to non-imprinted genes, and a repetitive element, Satellite 2 (referred to herein as SAT2CHRM1).


Significantly, this surprising result indicates that sperm abnormalities may be associated with a broad epigenetic defect of elevated DNA methylation at numerous sequences of diverse types, rather than a defect of imprinting alone as previously suggested [20].


To learn more about the possible extent of this apparent defect, the ILLUMINA™ platform was used to conduct DNA methylation analysis of 1,421 sequences in autosomal loci (discussed in more detail under Example 1 herein below). Briefly, the results of the ILLUMINA™ analyses appear in FIG. 3. Box 1 of FIG. 3 identifies 19 sequences with sperm-specific DNA methylation.


Various semen parameters have been correlated herein with abnormal DNA methylation (sperm concentration; total normal morphology; motility, volume, viscosity, etc.). According to preferred aspects, three of these semen parameters show the highest correlations with abnormal DNA methylation: sperm concentration; total normal morphology; and motility. FIG. 2, for example, shows that the corresponding MLL reactions are clustered based on sperm concentration.


Particular aspects of the present invention, therefore, provide marker(s) and marker subsets having utility for determining at least one of (A) abnormal sperm concentration, (B) abnormal morphology, and (C) abnormal motility. With respect to (A), abnormal sperm concentration, markers are provided in the following order of statistical significance from left to right, based on the p-value: HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, and CYP27B1. Nine of these markers have p-values well below 0.05, and therefore are very significant. Additionally provided are the markers, RNR1 and CYP27B1, both having p-values of 0.02, and therefore also provide for utility in this respect.


With respect to (B), abnormal total motile sperm, markers are provided in the following order of statistical significance from left to right, based on the p-value: HRAS, NTF3, MT1A (NTF3 and MT1A equally significant), SAT2CHRM1, DIRAS3, PLAGL1, MEST, PAX8, and SFN. These markers have p-values well below 0.05, and therefore are very significant. Additionally provided are the markers: RNR1 (p-value 0.04) and CYP27B1 and BDNF (both with p-value of 0.05), and therefore also provide for utility in this respect.


With respect to (C), abnormal motility, markers are provided in the following order of statistical significance from left to right, based on the p-value: MT1A, MEST, NTF3, PLAGL1. Additionally provided are the markers PAX8 AND ICAM1 (both having p-values of 0.05), and therefore also provide for utility in this respect.


Improper Erasure of Pre-Existing Methylation

According to particular aspects, only sequence-specific measures of DNA methylation are expected to reveal variation at individual sites, because of the enormous number of methylation targets in the human genome. These include millions of repetitive DNA elements for which methylation is postulated to silence parasitic and transposable activity. There are also large numbers of target sequences corresponding to single copy genes. Examples include thousands of promoter CpG islands for which methylation appears to mediate expression of genes in a tissue- and lineage-specific fashion, and DMRs associated with dozens of imprinted genes for which parent-of-origin DNA methylation marks are believed to mediate monoallelic expression in somatic cells.


As disclosed herein, Applicants' high-throughput analysis addressed hundreds of DNA methylation targets, and was thus designed to reveal methylation defects.


Elevated DNA methylation could, in theory, arise from either de novo methylation or improper erasure of pre-existing methylation. Although Applicants cannot rule out the possibility that processes responsible for de novo methylation are inappropriately activated in abnormal spermatogenesis, according to particular aspects, disruption of erasure is most likely the primary mechanism underlying abnormal spermatogenesis. Widespread erasure of DNA methylation occurs in both the pre-implantation embryo and again, uniquely, in primordial germ cells around the time that they enter the genital ridge. Several factors point to disruption of the second erasure as underlying the defect(s) described herein. Primordial germ cells arise from cells of the proximal epiblast which have themselves embarked upon somatic development, as shown by expression of somatic genes [25,26]. The germ cell lineage must therefore suppress the somatic program, which in mice is accomplished in part by genome-wide erasure of DNA methylation soon after germ cells migrate to the genital ridge [27]. This erasure affects DNA methylation on single copy genes, imprinted genes and repetitive elements [27]. Therefore, disruption of the second, genital ridge erasure most likely results in the type of pattern we observe in poor quality sperm, with elevated levels of DNA methylation at DNA sequences of each of these sequence types. Further, because this second erasure is confined to primordial germ cells, Applicants further reasoned that its disruption would be compatible with normal somatic development.


In humans, primordial germ cells colonize the genital ridge at about 4.5 weeks of gestation. Applicants are not aware of data describing DNA methylation in the human germ line at this date; however, the DMR in MEST at which Applicants found elevated DNA methylation in poor quality sperm is reportedly unmethylated in the male germ line by week 24 of gestation [28]. Potential causes of disrupted erasure have not been investigated. However, weeks 4.5-24 of gestation represent post-implantation stages of development wherein fetal physiology may be influenced by maternal factors and environmental compounds that cross the placenta. Possible origins of male infertility as early as 4.5 weeks of human gestation have not been studied. However, transient in vivo chemical exposure at 7-15 days post conception, which includes the analogous stage of murine development [29,30], results in spermatogenic deficits in rats with grossly normal testes [31] and may be associated with elevated methylation of sperm DNA [32].


Taken together, the observations disclosed herein indicate for the first time that epigenetic mechanisms contribute to a substantial portion of male factor infertility, and provide novel compositions and methods for the diagnosis, detection or determination of abnormal sperm or fertility. Also provided are methods for screening for agents that cause spermatogenic deficits, adbnormal sperm or fertility comprising: obtaining human ES-cell derived primordial germ cells; contacting the germ cells with at least one test agent; culturing the contacted germ cells; obtaining a sample of genomic DNA from the contacted cultured germ cells; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and identifying, based on the methylation status of the at least one CpG sequence, at least one test agent that causes spermatogenic deficits, adbnormal sperm or fertility.


Example 1
Sequence-Specific Levels of DNA Methylation were Related to Standard Measures of Sperm Quality
Overview

This is the first study ever to describe the epigenetic state of abnormal human sperm using an extensive panel of DNA methylation assays. To assess sperm DNA methylation at specific targets that are both more numerous and less specialized, a relatively larger set of sequence-specific assays was selected for use in the present study. DNA methylation was measured in ejaculated spermatozoa—interrogating sequences in repetitive elements, promoter CpG islands, and differentially methylated regions (DMRs) of imprinted genes. Then, to address the possible role of epigenetic programming in abnormal human spermatogenesis, sequence-specific levels of DNA methylation were related to standard measures of sperm quality.


Materials and Methods

Semen samples. Study semen samples were collected by 69 consecutive men ages 22-49 years who were partners of women undergoing evaluation for infertility at the Endocrine/Infertility Clinic of the Los Angeles County/University of Southern California Keck School of Medicine Medical Center. One additional semen sample was obtained from a sperm bank. The study was approved by the Institutional Review Board of the University of Southern California. Informed consent was not required because this research involved stored materials that had previously been collected solely for non-research purposes and were anonymous to the researchers/authors.


Semen Analysis. Standard semen analysis was performed using WHO criteria and Strict Morphology as previously described [33,34]. Semen volume, sperm concentration and motility, and leukocyte count were measured using the MicroCell chamber (Conception Technologies, San Diego, Calif.). Sperm morphology was assessed with the use of prestained slides (TestSimplets, Spectrum Technologies, Healdsburgh, Calif.), and percentage of morphologically normal sperm was documented. The samples were categorized according to concentration (<5, 5-20, >20 million sperm/ml), motility (<10, 10-50, >50 total motile sperm count (×106)), and morphology (<5%, 5-14%, >14% normal) of sperm [33,35]. Presence of any white blood cells, round cells, or epithelial cells was recorded. Following semen analysis, samples were stored at −30° C. until processing for molecular analysis.


Sperm Separation from Seminal Plasma. Semen samples were allowed to thaw at 37° C. Sperm were separated from seminal plasma using ISOLATE® Sperm Separation Medium (Irvine Scientific, Santa Ana, Calif.), a density gradient centrifugation column designed to separate cellular contaminants (including leukocytes, round cells, and miscellaneous debris) from spermatozoa [24]. Separation was performed according to the manufacturer's protocol [36], and the purity of separated sperm from contaminating cells was documented by light microscopy.


DNA isolation. DNA was isolated from purified sperm as previously described [37], with 0.1×SSC added to the Lysis buffer, and samples incubated at 55° C. over night or longer to complete the lysis procedure.


Laboratory Analysis of DNA Methylation. Sodium bisulfite conversion was performed as previously described [23]. The amount of DNA in each aliquot was normalized, and a bisulfite-dependent, DNA methylation-independent control reaction was performed to confirm relative amounts of DNA in each sample. METHYLIGHT™ analyses were performed as previously described [23]. Reaction IDs and sequences of the primers and probes used in the 294 METHYLIGHT™ reactions are as previously published (see Table 51 (Sections A-B): doi:10.1371/journal.pone.0001289.s001 (0.10 MB PDF; incorporated by reference herein in its entirety). Additionally, according to particular aspects of the present invention, names of preferred markers and respective primers, probes and genomic sequences corresponding to the respective amplicons are listed below in TABLE 1.









TABLE 1







Primers and Probes for exemplary preferred MethyLight Assays.
















Genomic sequence




Forward
Reverse
Probe Oligo
corresponding to



Primer
Primer
Sequence
amplicon Sequence


Gene
(SEQ ID NO:)
(SEQ ID NO:)
(SEQ ID NO:)
(SEQ ID NO:)





HRAS
GAGCGATGACG
CGTCCACAAAA
6FAM-
CGTCCACAAAATGGTTCTGG




GAATATAAGTT
TAATTCTAAAT
CACTCTTACCC
ATCAGCTGGATGGTCAGCGC



GG
CAACTAA
ACACCGCCGAC
ACTCTTGCCCACACCGCCGG



(SEQ ID NO: 46)
(SEQ ID NO: 47)
G-BHQ-1
CGCCCACCACCACCAGCTTA





(SEQ ID NO: 48)
TATTCCGTCATCGCTC






(SEQ ID NO: 20)





NTF3
TTTCGTTTTTG
CCGTTTCCGCC
6FAM-
CCCCGCCCTTGTATCTCATG



TATTTTATGGA
GTAATATTC
TCGCCACCACG
GAGGATTACGTGGGCAGCCC



GGATT
(SEQ ID NO: 29)
AAACTACCCAC
CGTGGTGGCGAACAGAACAT



(SEQ ID NO: 28)

G-BHQ-1
CACGGCGGAAACGG





(SEQ ID NO: 30)
(SEQ ID NO: 14)





MT1A
CGTGTTTTCGT
CTCGCTATCGC
6FAM-
CGTGTTCCCGTGTTACTGTG



GTTATTGTGTA
CTTACCTATCC
TCCACACCTAA
TACGGAGTAGTGGGTCCGAG



CG
(SEQ ID NO: 35)
ATCCCTCGAAC
GGACCTAGGTGTGGACAGGG



(SEQ ID NO: 34)

CCACT-BHQ-1
ACAGGCAAGGCGACAGCGAG





(SEQ ID NO: 36)
(SEQ ID NO: 16)





PAX8
CGGGATTTTTT
ACCTTTCCCCA
6 FAM-
CGGGACCTCCCTGTCGTACC



TGTCGTATTTG
TACTACCTCCG
ACGAACAATTC
TGAGAGGAGGGCCTGGCCCG



A
(SEQ ID NO: 26)
ACGAACCAAAC
TGAACTGCCCGTACACGGAG



(SEQ ID NO: 25)

CCTCCT-BHQ-1
GCAGCATGGGGAAAGGC





(SEQ ID NO: 27)
(SEQ ID NO: 13)





DIRAS3
GCGTAAGCGGA
CCGCGATTTTA
6 FAM-
GCGCAAGCGGAATCTATGCC



ATTTATGTTTG
TATTCCGACTT
CGCACAAAAAC
TGTTACCCACACTCCCTGCG



T
(SEQ ID NO: 32)
GAAATACGAAA
CCCCCGCACCCCGCTCCTGT



(SEQ ID NO: 31)

ACGCAAA-
GCGCAAGTCGGAATATAAAA





BHQ-1
CCGCGG





(SEQ ID NO: 33)
(SEQ ID NO: 15)





PLAGL1
ATCGACGGGTT
CTCGACGCAAC
6FAM-
ACCGACGGGCTGAATGACAA



GAATGATAAAT
CATCCTCTT
ACTACCGCGAA
ATGGCAGATGCCGTGGGCTT



G
(SEQ ID NO: 44)
CGACAAAACCC
TGCCGCCCGCGGCAGCCAAG



(SEQ ID NO: 43)

ACG-BHQ-1
AGGATGGCTGCGCCGAG





(SEQ ID NO: 45)
(SEQ ID NO: 19)





SFN
GAGGAGGGTTC
ATCGCACACGC
6FAM-
GAGGAGGGCTCGGAGGAGAA



GGAGGAGAA
CCTAAAACT
TCTCCCGATAC
GGGGCCCGAGGTGCGTGAGT



(SEQ ID NO: 40)
(SEQ ID NO: 41)
TCACGCACCTC
ACCGGGAGAAGGTGGAGACT





GAA-BHQ-1
GAGCTCCAGGGCGTGTGCGA





(SEQ ID NO: 42)
C






(SEQ ID NO: 18)





SAT2CHRM1
TCGAATGGAAT
CCATTCGAATC
6FAM-
TCGAATGGAATCAACATCCA



TAATATTTAAC
CATTCGATAAT
CGATTCCATTC
ACGGAAAAAAACGGAATTAT



GGAAAA
TCT
GATAATTCCGT
CGAATGGAATCGAAGAGAAT



(SEQ ID NO: 49)
(SEQ ID NO: 50)
TT-MGBNFQ
CATCGAATGGACCCGAATGG





(SEQ ID NO: 51)
(SEQ ID NO: 21)





MEST
CGGCGTTCGGT
CACACTCACCT
6FAM-
CGGCGCCCGGTGCTCTGCAA



GTTTTGTAA
ACGAAAACGAT
ACGCACCATAA
CGCTGCGGCGGGCGGCATGG



(SEQ ID NO: 37)
CTC
CATACC-BHQ-1
GATAACGCGGCCATGGTGCG




(SEQ ID NO: 38)
(SEQ ID NO: 39)
CCGAGATCGCCTCCGCAGGT






GAGTGTG






(SEQ ID NO: 17)





RNR1
CGTTTTGGAGA
AAACAACGCCG
6FAM-
CGCTCTGGAGACACGGGCCG



TACGGGTCG
AACCGAA
ACCGCCCGTAC
GCCCCCTGCGTGTGGCACGG



(SEQ ID NO: 52)
(SEQ ID NO: 53)
CACACGCAAA-
GCGGCCGGGAGGGCGTCCCC





BHQ-1
GGCCCGGCGCTGCTC





(SEQ ID NO: 54)
(SEQ ID NO: 22)





CYP27B1
GGGATAGTTAG
CCGAATATAAC
6FAM-
GGGACAGCCAGAGAGAACGG



AGAGAACGGAT
CACACCGCC
CCAACCTCAAC
ATGCCCATGAAATAAGGAAA



GTTT
(SEQ ID NO: 56)
TCGCCTTTTCC
AGGCGAGTTGAGGCTGGGGG



(SEQ ID NO: 55)

BHQ-1
CGGTGTGGCTACACTCGG





(SEQ ID NO: 57)
(SEQ ID NO: 23)





ICAM1
GGTTAGCGAGG
TCCCCTCCGAA
6FAM-
GGCCAGCGAGGGAGGATGAC



GAGGATGATT
ACAAATACTAC
TTCCGAACTAA
CCTCTCGGCCCGGGCACCCT



(SEQ ID NO: 58)
AA
CAAAATACCCG
GTCAGTCCGGAAATAACTGC




(SEQ ID NO: 59)
BHQ-1
AGCATTTGTTCCGGAGGGGA





(SEQ ID NO: 60)
(SEQ ID NO: 24)









Thirty-five METHYLIGHT™ reactions were selected for analysis of study sperm DNA samples based on cycle threshold (C(t)) values from analysis of the anonymous sample of sperm DNA. In brief, C(t) value is the PCR cycle number at which the emitted fluorescence is detectable above background levels. The C(t) value is inversely proportional to the amount of each methylated locus in the PCR reaction well, such that a low C(t) value suggests that the interrogated sequence is highly methylated. C(t) values of 35 or less were interpreted as an indication that a given sequence was methylated in the anonymous sample and selected 33 reactions on this basis. Three additional reactions were included, for which C(t) values slightly exceeded 35. Two (CYP27B1 and HOXA10) were selected based on gene function potentially related to fertility, and one (a non-CpG island reaction for IFNG) based on prior observation by applicants of hypomethylation in tumor versus normal tissue. When multiple reactions for a single locus resulted in C(t) values of less than 35, we selected only the reaction with the lowest C(t) value. Results of METHYLIGHT™ analysis were scored as PMR values as previously defined [23]. Following METHYLIGHT™ analyses, DNA remained from a subset of abnormal samples with greater sperm concentration. ILLUMINA™ analysis was performed on sodium bisulfite-converted sperm DNA of selected remaining samples, the anonymous semen sample, and purchased buffy coat DNA (HemaCare® Corporation, Van Nuys, Calif.) at the USC Genomics Core. Sodium bisulfite conversion for ILLUMINA™ assay was performed using the EZ-96 DNA Methylation Kit™ (ZYMO Research) according to manufacturer's protocol. Illumina Methods and reagents are as previously described [38]. The primer names and probe IDs are listed as previously published (see Table S2; doi:10.1371/journal.pone.0001289.s002 (0.20 MB PDF; incorporated by reference herein in its entirety), identifying 1,421 autosomal sequences of the GoldenGate Methylation Cancer Panel I, more fully described elsewhere [39,40]. Results of ILLUMINA™ assays were scored as β-values [38]. Relevant amplicons and CpG islands are provided below in TABLE 2 below.


Statistical association analyses of METHYLIGHT™ data. Associations between the ranked METHYLIGHT™ data and categorized semen values (Table 1) were tested using simple linear regression, with the semen characteristic categories scored as 0: low, 1: mid, 2: high. For selected sequences, boxplots of the methylation values (on the log(PMR+1) scale) are shown in FIG. 1. The top and bottom of the box denote the 75th and 25th percentiles, and the white bar the median. Whiskers are drawn to the observation farthest from the box that lies within 1.5 times the distance from the top to the bottom of the box, with values falling outside the whiskers denoted as lines. Results of this analysis were included in FIG. 1 for sequences associated with sperm concentration using the Benjamini and Hochberg procedure [41] to control the false discovery rate at 5%.


Statistical cluster analysis of METHYLIGHT™ data. Hierarchical cluster analysis of 36 loci was performed, using correlation to measure the distance between any two loci and Ward's method of linkage [42]. SASH1 was omitted from the cluster analysis because only a single sample showed positive methylation. The 65 study samples were ordered from left to right by increasing semen concentration.









TABLE 2





Exemplary, preferred amplicons and CpG islands






















HUGO








Gene


Reaction
Nomen-
Previously
Source of
UniGene
Reaction
Alternate Gene


Number
clature
Published?
published reaction
Number
ID
Name





HB-144
HRAS
Yes
Widschwendter, M.
Hs.37003
H-HRAS-
V-Ha-ras Harvey rat





et al Cancer Res

M1B
sarcoma viral





64, 3807-3813


oncogene homolog





(2004)


(HRAS); HRAS1


HB-251
NTF3
Yes
Weisenberger, D. J.
Hs.99171
H-NTF3-
Neurotrophin 3





et al Nature Genet

M1B





38, 787-793





(2006).


HB-205
MT1A
Yes
Weisenberger, D. J.
Hs.655199
H-MT1A-
Metallothionein





et al Nature Genet

M1B
1A/Metallothionein-I





38, 787-793





(2006).


HB-212
PAX8
No

Hs.469728
H-PAX8-
Paired Box Gene 8/







M3B
PAX8, Paired








Domain Gene 8,








PPARG Fusion








Gene


HB-043
DIRAS3
Yes
Fiegl, H. et al
Hs.194695
H-DIRAS3-
Ras homolog gene





Cancer Epidemiol

M1B
family, member





BioMark Prev


I/NOEY2; DIRAS





13, 882-888 (2004)


family, GTP-binding








RAS-like 3 (ARHI)


HB-199
PLAGL1
Yes
Weisenberger, D. J.
Hs.444975
H-
Pleiomorphic





et al Nature Genet

PLAGL1-
adenoma gene-like





38, 787-793

M1B
1/LOT1/Zac1





(2006).


HB-174
SFN
Yes
Weisenberger, D. J.
Hs.523718
H-SFN-
Stratifin/14-3-3





et al Nature Genet

M1B
protein sigma





38, 787-793





(2006).


HB-289
SAT2CHRM1
Yes
Weisenberger, D. J.
N/A
H-
SATELLITE 2





et al Nucleic Acids

SAT2CHRM1-
CHROMOSOME 1





Res 33, 6823-6836

M1M





(2005)


HB-493
MEST
No

Hs.270978
H-MEST-
PEG1







M2B


HB-071
RNR1
Yes
Muller, H. M. et al.
N/A
H-RNR1-
Ribosomal RNA





Cancer Lett209,

M1B





231-236 (2004)


HB-076
ICAM1
Yes
Ehrlich, M. et al.
Hs.643447
H-
Intercellular





Oncogene 21,

ICAM1B-
adhesion molecule 1





6694-6702 (2002)

M1B
(CD54), human








rhinovirus receptor


HB-223
CYP27B1
Yes
Weisenberger, D. J.
Hs.524528
H-
cytochrome P450,





et al Nature Genet

CYB27B1-
family 27, subfamily





38, 787-793

M1B
B, polypeptide 1





(2006).


















HUGO

GenBank


Length
Transcription



Gene

Accession
mRNA

of
Start


Reaction
Nomen-
Chromosomal
Number;
accession
Parallel/
Sequence
(GenBank


Number
clature
Location
version
number
Antiparallel
(bp)
Numbering)





HB-144
HRAS
11p15.5
AC137894;
NM_176795;
Antiparallel
165000
157238





AC137894.5
NM_176795.3





GI: 29650323
GI: 194363760


HB-251
NTF3
12p13
AC135585;
NM_002527;
Parallel
35700
7048





AC135585.2
NM_002527.4





GI: 24371348
GI: 156630993


HB-205
MT1A
16q13
AC106779;
NM_005946;
Parallel
158297
18787





AC106779.3
NM_005946.2





GI: 18483433
GI: 71274112


HB-212
PAX8
2q12
AC016683;
S77905;
Antiparallel
179937
116171





AC016683.7
S77905.1





GI: 11136842
GI: 998702


HB-043
DIRAS3
1p31
AF202543;
U96750;
Parallel
7242
2053





AF202543.1
U96750.1





GI: 11493727
GI: 4100354





(NG_011753.1
(NM_004675.2





GI: 226053757)
GI: 58530880)


HB-199
PLAGL1
6q24-q25
AL109755;
U72621;
Antiparallel
89669
53085





AL109755.14
U72621.4





GI: 6165524
GI: 21930302





(NG_009384.1
(Variants 1-8)





GI: 221136778(


HB-174
SFN
1p35.3
AF029081;
BC023552;
Parallel
10034
8563





AF029081.1
BC023552.2





GI: 2702352
GI: 33873404


HB-289
SAT2CHRM1
1
X72623;
N/A;
Parallel
1352
N/A





X72623.1





GI: 599824


HB-493
MEST
7q32.2
NC_000007;
NM_177524;
Parallel
20084
5893





NC_000007.13
NM_177524.1





GI: 224589819
GI: 29294634


HB-071
RNR1
13p12
X01547;
N/A
Parallel
850
482





X01547.1





GI: 35916


HB-076
ICAM1
19p13.3-p13.2
AC011511;
BC015969;
Parallel
156503
85732





AC011511.12
BC015969.2





GI: 21747446
GI: 33869582


HB-223
CYP27B1
12q14.1
AY288916;
AB005038;
Parallel
7587
1324





AY288916.1
AB005038.1





GI: 30527185
GI: 2626736






















Amplicon
Amplicon








Start
End
Mean







Location
Location
Distance







Relative to
Relative to
from





Amplicon
Amplicon
Transcription
Transcription
Transcription




HUGO
Location
Location
Start
Start
Start




Gene
Start
End
(bp,
(bp,
(bp,



Reaction
Nomen-
(GenBank
(GenBank
GenBank
GenBank
GenBank



Number
clature
Numbering)
Numbering)
sequence)
Sequence)
sequence)







HB-144
HRAS
156015
155920
1223
1318
1271



HB-251
NTF3
7503
7576
455
528
492



HB-205
MT1A
18175
18254
−612
−533
−573



HB-212
PAX8
72708
72632
43463
43539
43501



HB-043
DIRAS3
1953
2038
−100
−15
−58



HB-199
PLAGL1
53045
52969
40
116
78



HB-174
SFN
8848
8928
285
365
325



HB-289
SAT2CHRM1
1074
1153
N/A
N/A
N/A



HB-493
MEST
6057
6144
164
251
207



HB-071
RNR1
219
293
−263
−189
−226



HB-076
ICAM1
85597
85676
−135
−56
−96



HB-223
CYP27B1
1728
1805
404
481
443


















HUGO
Amplicon
Amplicon


Location of



Gene
Location Start
Location End
UCSC
UCSC
Amplicon in Gene


Reaction
Nomen-
(UCSC
(UCSC
Strand
Assembly
(e.g., promoter,


Number
clature
Numbering)
Numbering)
(+/−)
Date
exon)





HB-144
HRAS
524232
524327
+
May 2004
Exon2


HB-251
NTF3
5473982
5474055
+
May 2004
Exon1


HB-205
MT1A
55229471
55229550
+
May 2004
Promoter


HB-212
PAX8
113709183
113709259
+
May 2004
Exon 9


HB-043
DIRAS3
68228349
68228434

May 2004
Promoter (in Exon3)


HB-199
PLAGL1
1443711135
144371211
+
May 2004
Exon1


HB-174
SFN
26874056
26874136
+
May 2004
Exon1


HB-289
SAT2CHRM1
no perfect
no perfect match

May 2004
N/A




match


HB-493
MEST
129919339
129919425
+
March 2006
exon1/intron1


HB-071
RNR1
N/A
N/A

May 2004
Promoter


HB-076
ICAM1
10242630
10242709
+
May 2004
Promoter


HB-223
CYP27B1
56446731
56446808

May 2004
Exon1


















500 (approx. ±








250) bp




sequence


Location of
Location of




comprising

Estimated CpG Island
CpG Island
CpG Island



HUGO Gene
amplicon
CpG
Length (GenBank)
Start
End


Reaction
Nomen-
(Genbank
Island
(SEQ ID NO:)
(GenBank
(GenBank


Number
clature
sequence)
yes/no
(>0.6 CpG:GpC)
numbering)
numbering)





HB-144
HRAS
155726-156225
(Yes)
3354 (SEQ ID NO: 63)
156171
159524


HB-251
NTF3
7301-7800
Yes
 609 (SEQ ID NO: 2)
7246
7854


HB-205
MT1A
18201-18700
Yes
1209 (SEQ ID NO: 4)
17842
19050


HB-212
PAX8
72426-72925
Yes
1250 (SEQ ID NO: 1)
73859
72610


HB-043
DIRAS3
1751-2250
Yes
 552 (SEQ ID NO: 3)
1804
2355


HB-199
PLAGL1
52751-53250
Yes
1478 (SEQ ID NO: 7)
53667
52190


HB-174
SFN
8637-9136
Yes
 661 (SEQ ID NO: 6)
8684
9344


HB-289
SAT2CHRM1
 851-1350
(Yes)

(500 (SEQ ID NO: 9))

N/A
N/A


HB-493
MEST

Yes
2799 (SEQ ID NO: 5)
4293
7091


HB-071
RNR1
 1-500
yes
 850 (SEQ ID NO: 10)
1
850


HB-076
ICAM1
85376-85875
Yes
2038 (SEQ ID NO: 12)
84047
86084


HB-223
CYP27B1
1501-2000
yes
 747 (SEQ ID NO: 11)
1345
2091

















HUGO
Amplicon

Bisulfite




Gene
Start

Conversion:



Reaction
Nomen-
relative to

Top/Bottom



Number
clature
CGI start
Reaction Type
Strand







HB-144
HRAS
N/A
Methylated
Bottom



HB-251
NTF3
257
Methylated
Top



HB-205
MT1A
333
Methylated
Top



HB-212
PAX8
1151
Methylated
Top



HB-043
DIRAS3
149
Methylated
Top



HB-199
PLAGL1
622
Methylated
Top



HB-174
SFN
116
Methylated
Top



HB-289
SAT2CHRM1
N/A
Methylated
Top



HB-493
MEST
1764
Methylated
Top



HB-071
RNR1
219
Methylated
Top



HB-076
ICAM1
1685
Methylated
Top



HB-223
CYP27B1
383
Methylated
Top










Display of ILLUMINA™ data. ILLUMINA™ data were displayed graphically in FIG. 3 with results for study samples ordered left to right in columns by sperm concentration. Rows corresponding to each of the 1,421 sequences were divided into three tertiles of median β-value among buffy coat DNA samples (I, II, III), then sorted within tertile by median β-value among all sperm DNA samples. Box 1 contains all sequences tertile I with median β-value among sperm DNA samples >0.5; box 2 contains all sequences within tertile III with median β-value among sperm DNA samples <0.1. Maternal or paternal imprinting status of each locus was scored according to the categorization of R. Jirtle [43]. All sequences specific to genes imprinted in humans were individually reviewed to determine whether they have been reported as belonging to a DMR for which parent of origin marks are maintained by DNA methylation [44-66]. Sequences meeting these criteria were scored as maternally imprinted (MI) or paternally imprinted (PI) with an indicator set for each on FIG. 3.


Results

Standard semen analysis was conducted on samples collected by 69 men during clinical evaluation of couples with infertility. Among the 69 samples, semen volume ranged from 0.5 to 7.8 ml; total count 0 to 864 million sperm; total motile count 0 to 396.3 million sperm; and percentage normal sperm forms 0 to 26%. Four samples were found to be azoospermic and excluded from subsequent analysis of DNA methylation.


Applicants evaluated 294 METHYLIGHT™ reactions for the presence of methylation in sperm DNA from an anonymous semen sample obtained from a sperm bank. Primers and probes were as previously published (see Table 51 (Sections A-B), found at doi:10.1371/journal.pone.0001289.s001 (0.10 MB PDF); incorporated by reference herein in its entirety; Primers, probes and reaction IDs for 294 MethyLight Assays: Group A, used in screening procedure and analysis of 65 study samples; Group B, used only in screening procedure; and Group C, new assays designed to DMRs of maternally imprinted genes and used only in analysis of 65 study samples.


The 35 selected reactions of Table 51A were used to assay sperm DNA from 65 study samples.


At many of the 35 sequences methylation levels were elevated in DNA from poor quality sperm. For example, striking associations with each of sperm concentration, motility and morphology were observed for five sequences: HRAS, NTF3, MT1A, PAX8 and PLAGL1 (FIG. 1).


PLAGL1 is maternally imprinted. Our METHYLIGHT™ assay for this gene interrogates a differentially methylated CpG island [22]. To determine whether other maternally imprinted genes are methylated in abnormal sperm, METHYLIGHT™ was used to interrogate the differentially methylated sequence of DIRAS3. At this sequence greater DNA methylation was also observed in samples with poorer semen parameters (FIG. 1, row 6). These results appeared to conflict with those of Marques et al [20] who reported no association between low sperm count and methylation of a DMR in a third maternally imprinted gene, MEST. We therefore used METHYLIGHT™ to assess the methylation status of a differentially methylated MEST sequence investigated by these authors [20], and found elevated DNA methylation to be significantly associated with poor semen parameters (FIG. 1), in agreement with our PLAGL1 and DIRAS3 results.


After correction for multiple comparisons, estimated associations between results of each of the 37 METHYLIGHT™ assays and sperm concentration were highly significant for HRAS, NTF3, MT1A, PAX8, DIRAS3 and PLAGL1 and marginally significant for SFN, SAT2CHRM1 and MEST (Table 3, FIG. 1).









TABLE 3







Trend p-values for associations between MethyLight results and semen


parameters (see Methods).









Parameter of Standard Semen Analysis










MethyLight Reaction
Concentration
Motility
Morphology













*HRAS.HB.144
0.00006
0.00001
0.06265


*NTF3.HB.251
0.00029
0.00026
0.00464


MT1A.HB.205
0.00048
0.00026
0.00119


*PAX.8.HB.212
0.00086
0.00405
0.05143


*DIRAS3.HB.043
0.00109
0.00159
0.06016


*PLAGL1.HB.199
0.00213
0.00255
0.01951


*SFN.HB.174
0.00307
0.00804
0.79899


*SAT2CHRM1.HB.289
0.00448
0.00109
0.06793


*MEST.HB.493
0.00711
0.00373
0.00359


RNR1.HB.071
0.02
0.04
0.89


CYP27B1
0.02
0.05
0.10


MADH3.HB.053
0.09
0.15
0.35


BDNF.HB.257
0.11
0.05
0.26


PSEN1.HB.263
0.16
0.27
0.81


CGA.HB.237
0.23
0.34
0.93


SERPINB5.HB.208
0.23
0.64
0.80


ICAM1.HB.076
0.24
0.29
0.05


MINT1.HB.161
0.24
0.60
0.34


PTPN6.HB.273
0.24
0.09
0.08


ALU.HB.296
0.25
0.29
0.87


CYP1B1.HB.239
0.28
0.42
0.61


SP23.HB.301
0.28
0.48
0.48


IFNG.HB.311
0.33
0.22
0.93


C9.HB.403
0.37
0.35
0.89


GP2.HB.400
0.41
0.39
0.94


GATA4.HB.325
0.45
0.20
0.12


UIR.HB.189
0.48
0.47
0.70


TFF1.HB.244
0.48
0.96
0.93


LDLR.HB.219
0.51
0.39
0.11


SASH1.HB.085
0.51
0.15
0.15


ABCB1.HB.051
0.54
0.27
0.16


HOXA10.HB.270
0.63
0.84
0.13


MTHFR.HB.058
0.70
0.38
0.43


LINE1.HB.330
0.87
0.47
0.14


LZTS1.HB.200
0.90
0.95
0.73


SMUG1.HB.086
0.90
0.36
0.76



IGF2.HB.345

0.91
0.71
0.11





*Belongs to cluster 2 (see FIG. 2).



Assay interrogates a non-differentially methylated sequence.



Trends were assessed over the following categories of semen parameters: Concentration (<5, 5-20, >20 × 106 sperm per ml), Morphology (<5%, 5-14%, >14% normal sperm forms), Motility (<10, 10-50, >50 total motile sperm count (×106)).






Applicants then subjected METHYLIGHT™ data from 36 of the assays to unsupervised cluster analysis. (Data for SASH1 were not included, because methylation at this sequence was detected in only one sample.) This analysis identified three distinct clusters of sequences based on DNA methylation profiles in the 65 samples (FIG. 2). Notably, the middle cluster shown in FIG. 2 includes eight of the nine sequences (all except MT1A) individually associated with semen parameters. This middle cluster includes not only three sequences that are differentially methylated on imprinted loci, but also three single copy sequences specific to non-imprinted genes, and a repetitive element, Satellite 2 [23] (reaction named SAT2CHRM1).


Significantly, this surprising result indicates that sperm abnormalities may be associated with a broad epigenetic defect of elevated DNA methylation at numerous sequences of diverse types, rather than a defect of imprinting alone as previously suggested [20].


To learn more about the possible extent of this apparent defect, the ILLUMINA™ platform was used to conduct DNA methylation analysis of 1,421 sequences in autosomal loci. Included in this analysis was: DNA from the anonymous sperm sample used in the METHYLIGHT™ screen (FIG. 3, columns S); two purchased samples of buffy coat DNA allowing for observation of methylation patterns in somatic cells (FIG. 3, columns 1-2), and seven study sperm DNA samples remaining after METHYLIGHT™ analysis (FIGS. 2-3, columns A-G).


Results of ILLUMINA™ analyses appear in FIG. 3. A large number of genes were similarly methylated in both sperm DNA and buffy coat DNA (blue regions on the left bar, I; red regions on the right bar, III), while others tended to be more methylated in DNA isolated from only one of these cell types. Boxes enclose sequences for which we observed particularly strong patterns of cell type-specific methylation. Box 1 identifies 19 sequences with sperm-specific DNA methylation. At these sequences, methylation profiles of all DNA from samples of study sperm (A-G) closely resemble those from the anonymous sperm sample and differ greatly from those of buffy coat DNA. Box 2 identifies 102 sequences with buffy coat-specific DNA methylation. This set is larger in number than the sperm-specific set, as expected, given that sperm DNA is reportedly hypomethylated compared with somatic cell DNA [14]. The buffy coat-specific set comprises 7.2% of the 1,421 sequences including the majority of DMRs associated with imprinted genes that are on the Illumina panel. At many buffy coat-specific sequences, DNA methylation was elevated in study sperm DNA, most notably in sample A that had been isolated from sperm with the lowest concentration among samples A-G. Methylation of sample A DNA is elevated (β>0.1) at 76 of the 102 sequences in box 2, including all 10 that are known DMRs associated with imprinted genes.


Several factors assure us that our observations did not arise from somatic cell contamination of separated sperm samples [21]. Somatic cells are far larger than sperm and readily identified by microscopic evaluation of semen samples. Even if somatic cells are present in the neat ejaculate, the ISOLATE® sperm separation technique is specifically designed to separate spermatozoa from somatic cells and miscellaneous debris [24]. Moreover, although microscopic evaluation of semen samples conducted before sperm separation identified white blood cells in five of the 65 neat semen samples, excluding results on these five samples from statistical analyses had minimal effect on associations between DNA methylation and semen parameters, and DNA from these samples were excluded from ILLUMINA™ assays.


Various semen parameters have been correlated with abnormal DNA methylation (sperm concentration; total normal morphology; motility, volume, viscosity, etc.). According to preferred aspects, three of these semen parameters show the highest correlations with abnormal DNA methylation: sperm concentration; total normal morphology; and motility. FIG. 2, for example, shows that the corresponding MLL reactions are clustered based on sperm concentration.


Particular preferred aspects, therefore, provide marker(s) and marker subsets having utility for determining at least one of abnormal sperm concentration, abnormal morphology, and abnormal motility.


In particular aspects, with respect to (A) abnormal sperm concentration, markers are provided in the following order of statistical significance from left to right, based on the p-value: HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, and MEST. All of these nine markers have p-values well below 0.05, and therefore, all nine are very significant. Additionally provided are two more markers, RNR1 and CYP27B1, both have p-value of 0.02, that are therefore also of utility in this respect.


In particular aspects, with respect to (B) abnormal total motile sperm, markers are provided in the following order of statistical significance from left to right, based on the p-value: HRAS, NTF3, MT1A (NTF3 and MT1A equally significant), SAT2CHRM1, DIRAS3, PLAGL1, MEST, PAX8, & SFN. Again, these have very significant p-values. Additionally provided are three more markers: RNR1(p-value 0.04) and CYP27B1, BDNF, both with p-value of 0.05, that are therefore also of utility in this respect.


In particular aspects, with respect to (C) abnormal motility, markers are provided in the following order of statistical significance from left to right, based on the p-value: MT1A, MEST, NTF3, PLAGL1. Additionally, PAX8 AND ICAM1 both have p-values of 0.05, and are thus also of utility in this respect.


Example 2
Additional Aspects Provide Methods for Screening for Agents that Cause Spermatogenic Deficits, Abnormal Sperm or Abnormal Fertility
Overview

As stated herein above, this is the first study ever to describe the epigenetic state of abnormal human sperm using an extensive panel of DNA methylation assays. According to additional aspects, Applicants data has provided novel methylation-based markers for abnormal human sperm and/or fertility.


As recognized in the art, transient in vivo chemical exposure at 7-15 days post conception, which includes the analogous stage of murine development [29,30], results in spermatogenic deficits in rats with grossly normal testes [31] but likely associated with elevated methylation of sperm DNA [32].


According to additional aspects, therefore, Applicants' data provides for methods for screening for agents that cause spermatogenic deficits, abnormal sperm or abnormal fertility. In particular aspects, ES-cell derived primordial germ cells are exposed to chemical test agents, followed by CpG methylation analysis as described and provided for herein, to allow for a high-throughput screening assay to test and identify agents that cause spermatogenic deficits, abnormal sperm or abnormal fertility. Culturing of embryonic stem (ES) cells to efficiently provide for primordial germ cells is known in the art. For example, human embryonic stem (ES) cells are propagated on mouse embryo fibroblast feeder cells as described (67). A multistep induction procedure incorporating several previously described protocols can be used to convert ES cells into primordial germ cells at high efficiency. For example, ES cells are treated with bone morphogenetic protein-2 for a brief 24 period in combination with activin and FGF-2 in chemically defined medium. After 24 hours the BMP-2 is removed and retinoic acid is added. As will be appreciated in the art, a range of doses of each factor may be employed in a matrix design over a variable time course to optimize the yield of c-kit positive/placental alkaline phosphatase positive cells. These cells are isolated by flow cytometry and subjected to Q-RTPCR to analyze for the presence of primordial germ cell and gonocyte specific genes such as VASA. According to particular aspects, up to 10% of the treated cells are vasa positive following optimal treatment. Primordial germ cells and gonocytes may also be isolated from embryonic and fetal gonads by the use of c-kit and placental alkaline phosphatase in combination with flow cytometry, following collagenase and Tryple Express™ digestion of the tissue.


Particular aspects, therefore, provide methods for screening for agents that cause spermatogenic deficits, abnormal sperm or abnormal fertility comprising: obtaining human ES-cell derived primordial germ cells; contacting the germ cells or descendants thereof, with at least one test agent; culturing the contacted germ cells or the descendants thereof under conditions suitable for germ cell proliferation or development; obtaining a sample of genomic DNA from the contacted cultured germ cells or the descendants thereof; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and identifying, based on the methylation status of the at least one CpG sequence, at least one test agent that causes at least one of spermatogenic deficits, abnormal sperm, and abnormal fertility. In certain embodiments, the determined methylation status of the at least one CpG sequence is hypermethylation. In preferred embodiments, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


Example 3
Additional Aspects Provide for Diagnosis and/or Prognosis of Testicular Cancer, Comprising Use of the Biomarkers Disclosed Herein

Overview. Testicular germ cell tumors are the most common cancer of young men. Platinum-based chemotherapeutic agents improved survival of these tumors dramatically in the 1970's, to nearly 80 percent; in response to this success, etiologic and preventive research waned. However, in the intervening decades, incidence has doubled, and it has become clear that patients cured of germ cell tumors have serious and sometimes fatal sequelae. One of the few demonstrated risk factors is primary infertility (preceding cancer diagnosis), and age-period-cohort analyses implicate events during embryogenesis or early childhood (reviewed in Cortessis 2003).


Germ cells have been called the “ultimate” stem cells because they can give rise to all cell types of the body. Germ cells and their transformed derivatives share many properties with


embryonic stem cells, including expression of OCT4 and Nanog transcription factors (Sperger et al 2003) required for maintenance of stem cell pluripotency (Boyer 2005), capacity for both differentiation and transformation during in vitro manipulation, and gain of supernumerary material of common regions of chromosomes 12 and 17 (Draper 2004).


The mammalian germ line undergoes extensive epigenetic reprogramming during development. Widespread erasure of DNA methylation occurs in primordial germ cells as they migrate to the gonadal ridges (FIGS. 4a and b). After the testes form, de novo DNA methylation (FIG. 4b) establishes male-specific DNA methylation during germ cell maturation and spermatogenesis (Hajkova 2002). Consistent with the possibility that disruption of epigenetic reprogramming of the germ line leads to abnormal spermatogenesis and compromised male fertility, we (Houshdaran et al 2007) and others (Marques 2008) demonstrated DNA hypermethylation in sperm of men with poor quality sperm, a clinical indicator of sub-fertility.


According to the cancer stem cell hypothesis of carcinogenesis, malignant tumors include cells with the stem cell properties of self-renewal and pluripotency. This hypothesis addresses demonstrated features of stem cells and tumors, accounts in part for the transformed phenotypes and evolving behavior of malignancies, and largely explains the distribution of malignant potential among the tissue types of the body.


According to particular aspects, if the general hypothesis is correct, the rapidly emerging principles and tools of stem cell biology will likely have tremendous potential to shed light on specific mechanisms of carcinogenesis, and to provide molecular strategies for cancer prevention and treatment. However, research addressing the hypothesis requires the identification of stem cells within the tumor, an objective not yet attained for most human malignancies. Fortunately, this obstacle does not exist for germ cell tumors, for which the stem cell component is the germ cells themselves, which are readily identified.


As discussed herein, epigenetic change is a feature shared by normally differentiating stem cells and abnormally evolving cancer cells. The germ cell lineage is extensively reprogrammed during embryonic development, and the epigenetic state of germ cells is particularly susceptible to disruption during this time. Epidemiologic studies have consistently identified this same period as critical in germ cell carcinogenesis, and shown that infertility is associated with risk of germ cell tumors.


According to additional aspects of the present invention, disruption of epigenetic programming of germ cells during early development leads to both functional deficits manifest as sub-fertility in adulthood and increased malignant potential.


As described herein under Applicants' Examples 1 and 2, we first examined DNA methylation of sperm provided by men with poor fertility—a more common condition for which samples could be readily obtained. Our results disclosed herein are consistent with Applicants' additional conception, as they can be explained by disruption of the widespread erasure of DNA methylation that occurs early in the development of germ cells (see FIG. 4). FIGS. 4a, b and c schematically depict mammalian primordial germ line development, showing that primordial germ cells undergo extensive epigenetic reprogramming during development, where widespread erasure of DNA methylation occurs in primordial germ cells as they migrate to the gonadal ridges). After the testes form, de novo DNA methylation establishes male-specific DNA methylation during germ cell maturation and spermatogenesis (see FIGS. 4a and b). According to particular aspects of the present invention, DNA methylation of sperm from men with poor fertility is consistent with disruption of the widespread erasure of DNA methylation that occurs early in the development of germ cells (see FIG. 4c).


DNA methylation analyses revealing epigenetic stem cell signature in epithelial cancers. In stem cells, polycomb group proteins reversibly repress genes required for differentiation. Using DNA methylation analysis of gene promoter sequences, polycomb target genes were recently found to be over-represented among genes that are silenced in cancers of the colon, ovary and breast by DNA hypermethylation of promoter sequences (Widschwendter et al 2007).


According to additional aspects of the present invention, the epigenetic state of stem cell components of human germ cell tumors and sub-fertile spermatozoa are compared with that of normal germ cell populations from which they originate to provide a set of biological markers for germ cell tumors and sub-fertile spermatozoa.


Materials and Methods:

Characterization of DNA methylation of cells representing very early and final stages of the male germ cell lineage (germ cell precursors and spermatozoa) identifying profiles that distinguish between these stages. Using an Illumina Infinium assay a quantitative assessment of DNA methylation is performed at over 25,000 DNA sequences in order to determine normal DNA methylation profiles in each cell type. By statistical analysis the sequences at which DNA methylation best distinguishes the epigenetic state of early versus late stages of normally developing male germ cells are identified.


Development and validation of MethyLight assays of DNA methylation at identified sequences. MethyLight assays are first be used to measure DNA methylation in the cell types used above, then to assess any cell types representing intermediate stages of development (e.g. gonocytes) for which fresh cells of sufficient quantity could not be. On the basis of these results, subsets of reactions representing probable stages of germ line development will be identified.


Assessment of DNA methylation of germ cells provided by men diagnosed with germ cell tumors and sub-fertility using the developed MethyLight assays. DNA of spermatozoa collected prior to treatment is assessed for patients diagnosed with both germ cell tumors and male-factor infertility. For germ cell tumor patients, germ cells taken from testicular tissue are also examined. For the set of patients for whom fresh tissue is collected at orchiectomy, germ cells from both tumor and normal regions of the testis are analyzed, while germ cell-rich tumors are analyzed, only, of specimens obtained as formalin-fixed paraffinembedded (FFPE) tissue. Use of FFPE tissue allows inclusion of a representative sample of histologic types of germ cell tumors, while analysis of the fresh tissue allows for separate examination of normal and transformed germ cells of the same patient. The resulting patterns are compared to those identified above, to confirm that aberrant epigenetic state of germ cell DNA is a component of the etiology of these conditions. These additional aspects comprise combining the knowledge and methods of cancer epidemiology, stem cell biology and epigenetics.


Etiologic studies of testicular germ cell tumors. Applicants have informed consent access to a collection of fresh testicular germ cell tumor tissue.


In vitro preparation of germ cell precursors. Human embryonic stem (ES) cells are propagated on mouse embryo fibroblast feeder cells as described (Reubinoff et al., 2000). Embryonal carcinoma cell lines are propagated as described previously (Pera et al., 1989). A multistep induction procedure incorporating several previously described protocols is used to convert ES cells into primordial germ cells at high efficiency. ES cells are treated with bone morphogenetic protein-2 for a brief 24 period in combination with activin and FGF-2 in chemically defined medium. After 24 hours the BMP-2 is removed and retinoic acid is added. A range of doses of each factor is employed in a matrix design over a variable time course to optimize the yield of c-kit positive/placental alkaline phosphatase positive cells. These cells are isolated by flow cytometry and subjected to Q-RTPCR to analyze for the presence of primordial germ cell and gonocyte specific genes such as VASA. Applicants estimate that up to 10% of cells will be vasa positive following optimal treatment.


Samples. Human ES and embryonal carcinoma cells are already available. Fresh testicular tissue representing normal embryonic, fetal, pre-pubescent and adult stages of development are available through an ongoing protocol of tissue collection at postmortem exam. Fresh testicular tissue from men diagnosed with germ cell tumors is collected at orchiectomy and made available. Paraffin-embedded formalin-fixed (PEFF) tumor blocks are obtainable from the discard repository of cancer surveillance programs and by requesting blocks to be prepared from tissue resected from patients who have provided consent for same. Normal sperm is taken from donated sperm bank samples and maintained in a research repository. Semen samples are collected by patients newly diagnosed with testicular germ cell tumors and infertility before treatment for either condition. To obtain semen samples from testis cancer patients, logistical procedures are used before initial treatment of testis cancer. Semen samples from infertility patients are available.


Sample processing. Primordial germ cells and gonocytes are isolated from embryonic and fetal gonads by the use of c-kit and placental alkaline phosphatase in combination with flow cytometry, following collagenase and Tryple Express digestion of the tissue. This protocol is evaluated for isolation of more mature germ cells and adapted by use of additional antibodies as required. Sperm is separated from seminal plasma as described (Houshdaran et al 2007). DNA is isolated from all cell types to be studied, and subjected to bisulfite conversion prior to analysis of DNA methylation.


Analysis of DNA methylation. Illumina Infinium Methylation assays are used, and which can generate DNA methylation data for 27,578 loci on sets of 12 samples at once, performing optimally using DNA isolated from fresh or fresh-frozen cells. The Infinium assay is used for samples for which abundant DNA can be isolated from fresh cells, and MethyLight assays for all other (fewer cells from fresh and all from FFPE tissue).


Statistical analysis. Analysis relating patterns of DNA methylation to each cell type and disease outcome is performed in collaboration with a mathematical statistician who specializes in statistical methods for analysis of DNA methylation data. Approximately 20 loci are identified for which DNA methylation levels, in combination, provide the highest sensitivity and specificity for distinguishing early from late cell types in the germ cell lineage. MethyLight assays are developed for these sequences, and DNA methylation profiles from germ cells of men with these conditions are compared systematically to the profiles of normal cells representing early and late stages of germ cell maturation using multivariate analysis implemented using machine learning algorithms.


Translational Potential. This Example provides a new combination of cancer epidemiology, stem cell biology, and epigenetics, and addresses multiple conditions ranging from infertility to several cancers.


Results. Abnormal DNA methylation of poor quality sperm. As described elsewhere herein, Applicants have identified a pattern of elevated DNA methylation in DNA of poor quality sperm (Houshdaran et al 2007). Notable features of the pattern are the association with a large proportion of interrogated sequences in multiple epigenetic contexts including promoter CpG islands of expressed genes, differentially methylated sequences of imprinted genes, and a repeated element, and a consistent dose response pattern of higher DNA methylation among samples with poorer values of each parameter of the standard semen analysis (see boxplots relating summary “M index” of MethyLight results at all associated sequences to individual semen parameters, FIG. 2). Of particular relevance to the subject matter of this Example were associations with sequences specific to cancer-related HRAS1 (p=0.00006), and NTF3 (p=0.00029) implicated in germ cell carcinogenesis based on protein function and presence in the region of chromosome 12p that is consistently amplified in malignant transformation of germ cells and embryonic stem cells.


Additional aspects, therefore, provide methods for diagnosis or prognosis of testicular or male germ-cell derived cancer, comprising: obtaining a sample of human sperm DNA from a test subject; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and determining, based on the methylation status of the at least one CpG sequence, diagnosis or prognosis of testicular or male germ-cell derived cancer with respect to the test subject. In certain aspects, the determined methylation status of the at least one CpG sequence is hypermethylation. In particular aspects, determining the methylation status of at least one CpG dinucleotide sequence comprises 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. In certain embodiments, treating comprises use of bisulfite treatment of the DNA. In particular aspects, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


In certain aspects, the diagnosis or prognosis is of germ-cell derived testicular cancer.


Particular embodiments of the cancer diagnostic or prognostic assays comprise determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8 and PLAGL1. In certain aspects, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, and PLAGL1 SEQ ID NOS:7 and 19.'


Additional aspects provide a method for diagnosis or prognosis of testicular or male germ-cell derived cancer, comprising: obtaining a sample of human sperm DNA from a test subject; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence from each of a repetitive DNA element sequence group, a maternally imprinted gene sequence group, and a non-imprinted gene sequence group; and determining, based on the methylation status of the at least one CpG sequence from each of the groups, diagnosis or prognosis of testicular or male germ-cell derived cancer with respect to the test subject. In certain aspects, the determined methylation status of the at least one CpG sequence is hypermethylation. In particular implementations, the methylation status of at least one CpG dinucleotide sequence comprises 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. In certain embodiments, treating comprises use of bisulfite treatment of the DNA. In certain aspects, the at least one gene sequence from a repetitive element group comprises at least one selected from the group consisting of SAT2CHRM1 SEQ ID NOS:9 and 21. In particular aspects, the at least one gene sequence from a maternally imprinted gene group comprises at least one selected from the group consisting of PLAGL1 SEQ ID NOS:7 and 19, MEST SEQ ID NOS:5 and 17, and DIRAS3 SEQ ID NOS:3 and 15. In certain embodiments, the at least one gene sequence from a non-imprinted gene group comprises at least one selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, SFN SEQ ID NOS:6 and 18, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.


Additionally provided are methods for screening for agents that cause testicular or male germ-cell derived cancer, comprising: obtaining human ES-cell derived primordial germ cells; contacting the germ cells or descendants thereof, with at least one test agent; culturing the contacted germ cells or the descendants thereof under conditions suitable for germ cell proliferation or development; obtaining a sample of genomic DNA from the contacted cultured germ cells or the descendants thereof; determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; and identifying, based on the methylation status of the at least one CpG sequence, at least one test agent that causes testicular or male germ-cell derived cancer. In certain aspects, the determined methylation status of the at least one CpG sequence is hypermethylation. In certain aspectgs, determining the methylation status of at least one CpG dinucleotide sequence comprises 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. In certain embodiments, treating comprises use of bisulfite treatment of the DNA. In particular embodiments, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24. In certain aspects, the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, and PLAGL1 SEQ ID NOS:7 and 19.


CITED REFERENCES, INCORPORATED HEREIN BY REFERENCE



  • Boyer L A. Lee T I. Cole M F. Johnstone S E. Levine S S. Zucker J P. Guenther M G. Kumar R M.

  • Murray H L. Jenner R G. Gifford D K. Melton D A. Jaenisch R. Young R A. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 122:947-56, 2005.

  • Cortessis V. Epidemiologic Insights into the Occurrence and Causes of Testicular Cancer In Raghavan D (ed), American Cancer Society Atlas of Clinical Oncology—Germ Cell Tumors, B C Decker Inc, Hamilton, London, 2003.

  • Draper J S, Smith K, Gokhale P, Moore H D, Maltby E, Johnson J. Recurrent gain of chromosomes 17q and 12 in cultured human embryonic stem cells. Nature Biotechnology 22: 53-4, 2004.

  • Hajkova P, Erhardt S, Lane N, Haaf T, El-Maarri O, Reik W, Walter J, Surani M A. Epigenetic reprogramming in mouse primordial germ cells. Mech Dev 117: 15-23, 2002.

  • Houshdaran S, Cortessis V, Siegmund K, Yang A, Laird P, Sokol R. Widespread epigenetic abnormalities suggest a broad DNA methylation erasure defect in abnormal human sperm. PloS ONE 2:e1289,2007.

  • Marques C J. Costa P. Vaz B. Carvalho F. Fernandes S. Barros A. Sousa M. Abnormal methylation of imprinted genes in human sperm is associated with oligozoospermia. Molecular Human Reproduction. 14:67-74, 2008.

  • Pera M F, Cooper S, Mills J, Parrington J M. Isolation and characterization of a multipotent clone of human embryonal carcinoma cells. Differentiation; research in biological diversity 42 10-23, 1989.

  • Reubinoff B E, Pera M F, Fong C Y, Trounson A, Bongso A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol 18 399-404, 2000.

  • Sperger J M, Chen X, Draper J S, Antosiewicz J E, Chon C H, Jones S B, et al. Gene expression patterns in human embryonic stem cells and human pluripotent germ cell tumors. PNAS 100: 13350-5, 2003.

  • Widschwendter M. Fiegl H. Egle D. Mueller-Holzner E. Spizzo G. Marth C. Weisenberger D J. Campan M. Young J. Jacobs I. Laird P W. Epigenetic stem cell signature in cancer. Nature Gnetics. 39:157-8, 2007.

  • 1. Sokol R Z (1997) male factor in male infertility. In: Lobo R. MD, Paulson R., editor. Infertility, Contraception, and Reproductive Endocrinology. Madden, Mass.: Blackwell Sciene, Inc. pp. 547-566.

  • 2. Thonneau P, Marchand S, Tallec A, Ferial M L, Ducot Bet al. (1991) Incidence and main causes of infertility in a resident population (1,850,000) of three French regions (1988-1989). Hum Reprod 6(6): 811-816.

  • 3. Maduro M R, Lo K C, Chuang W W, Lamb D J (2003) Genes and male infertility: what can go wrong? J Androl 24(4): 485-493.

  • 4. McElreavey K, Krausz C, Bishop C E (2000) The human Y chromosome and male infertility. Results Probl Cell Differ 28: 211-232.

  • 5. Sharlip I D, Jarow J P, Belker A M, Lipshultz L I, Sigman M et al. (2002) Best practice policies for male infertility. Fertil Steril 77(5): 873-882.

  • 6. Rousseaux S, Caron C, Govin J, Lestrat C, Faure A K et al. (2005) Establishment of male-specific epigenetic information. Gene 345(2): 139-153.

  • 7. Emery B R, Carrell D T (2006) The effect of epigenetic sperm abnormalities on early embryogenesis. Asian J Androl 8(2): 131-142.

  • 8. Li E (2002) Chromatin modification and epigenetic reprogramming in mammalian development. Nat Rev Genet 3(9): 662-673.

  • 9. Saitou M, Barton S C, Surani M A (2002) A molecular programme for the specification of germ cell fate in mice. Nature 418(6895): 293-300.

  • 10. Santos F, Dean W (2004) Epigenetic reprogramming during early development in mammals. Reproduction 127(6): 643-651.

  • 11. Biermann K, Steger K (2007) Epigenetics in Male Germ Cells. J. Androl.

  • 12. Ariel M, Cedar H, McCarrey J (1994) Developmental changes in methylation of spermatogenesis-specific genes include reprogramming in the epididymis. Nat Genet 7(1): 59-63.

  • 13. Trasler J M (1998) Origin and roles of genomic methylation patterns in male germ cells. Semin Cell Dev Biol 9(4): 467-474.

  • 14. Oakes C C, La Salle S, Smiraglia D J, Robaire B, Trasler J M (2007) A unique configuration of genome-wide DNA methylation patterns in the testis. Proc Natl Acad Sci USA 104(1): 228-233.

  • 15. Bestor T H, Bourc'his D (2004) Transposon silencing and imprint establishment in mammalian germ cells. Cold Spring Harb Symp Quant Biol 69: 381-387.

  • 16. Schaefer C B, Ooi S K, Bestor T H, Bourc'his D (2007) Epigenetic decisions in mammalian germ cells. Science 316(5823): 398-399.

  • 17. Flanagan J M, Popendikyte V, Pozdniakovaite N, Sobolev M, Assadzadeh A et al. (2006) Intra- and interindividual epigenetic variation in human germ cells. Am J Hum Genet 79(1): 67-84.

  • 18. Benchaib M, Braun V, Ressnikof D, Lornage J, Durand P et al. (2005) Influence of global sperm DNA methylation on IVF results. Hum Reprod 20(3): 768-773.

  • 19. Hartmann S, Bergmann M, Bohle R M, Weidner W, Steger K (2006) Genetic imprinting during impaired spermatogenesis. Mol Hum Reprod 12(6): 407-411.

  • 20. Marques C J, Carvalho F, Sousa M, Barros A (2004) Genomic imprinting in disruptive spermatogenesis. Lancet 363(9422): 1700-1702.

  • 21. Manning M, Lissens W, Liebaers I, Van Steirteghem A, Weidner W (2001) Imprinting analysis in spermatozoa prepared for intracytoplasmic sperm injection (ICSI). Int J Androl 24(2): 87-94.

  • 22. Varrault A, Bilanges B, Mackay D J, Basyuk E, Ahr B et al. (2001) Characterization of the methylation-sensitive promoter of the imprinted ZAC gene supports its role in transient neonatal diabetes mellitus. J Biol Chem. pp. 18653-18656.

  • 23. Weisenberger D J, Siegmund K D, Campan M, Young J, Long T I et al. (2006) CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 38(7): 787-793.

  • 24. Dale B, Elder, K. (1997) In vitro fertilization. New York: Cambridge University Press. 187 p.

  • 25. Hayashi K, de Sousa Lopes S M, Surani M A (2007) Germ cell specification in mice. Science 316(5823): 394-396.

  • 26. Yabuta Y, Kurimoto K, Ohinata Y, Seki Y, Saitou M (2006) Gene expression dynamics during germline specification in mice identified by quantitative single-cell gene expression profiling. Biol Reprod 75(5): 705-716.

  • 27. Surani M A, Hayashi K, Hajkova P (2007) Genetic and epigenetic regulators of pluripotency. Cell 128(4): 747-762.

  • 28. Kerjean A, Dupont J M, Vasseur C, Le Tessier D, Cuisset L et al. (2000) Establishment of the paternal methylation imprint of the human H19 and MEST/PEG1 genes during spermatogenesis. Hum Mol Genet 9(14): 2183-2187.

  • 29. Lee J, Inoue K, Ono R, Ogonuki N, Kohda T et al. (2002) Erasing genomic imprinting memory in mouse clone embryos produced from day 11.5 primordial germ cells. Development 129(8): 1807-1817.

  • 30. Hajkova P, Erhardt S, Lane N, Haaf T, El-Maarri O et al. (2002) Epigenetic reprogramming in mouse primordial germ cells. Mech Dev 117(1-2): 15-23.

  • 31. Cupp A S, Uzumcu M, Suzuki H, Dirks K, Phillips B et al. (2003) Effect of transient embryonic in vivo exposure to the endocrine disruptor methoxychlor on embryonic and postnatal testis development. J Androl 24(5): 736-745.

  • 32. Chang H S, Anway M D, Rekow S S, Skinner M K (2006) Transgenerational epigenetic imprinting of the male germline by endocrine disruptor exposure during gonadal sex determination. Endocrinology 147(12): 5524-5541.

  • 33. Acacio B D, Gottfried T, Israel R, Sokol R Z (2000) Evaluation of a large cohort of men presenting for a screening semen analysis. Fertil Steril 73(3): 595-597.

  • 34. World Health Organization Laboratory Manual for Human Semen and Sperm Cervical Mucus Interaction (1999).

  • 35. Guzick D S, Overstreet J W, Factor-Litvak P, Brazil C K, Nakajima S T et al. (2001) Sperm morphology, motility, and concentration in fertile and infertile men. N Engl J Med 345(19): 1388-1393.

  • 36. www.irvinesci.com (2006).

  • 37. Laird P W, Zijderveld A, Linders K, Rudnicki M A, Jaenisch R et al. (1991) Simplified mammalian DNA isolation procedure. Nucleic Acids Res 19(15): 4293.

  • 38. Bibikova M, Lin Z, Zhou L, Chudin E, Garcia E W et al. (2006) High-throughput DNA methylation profiling using universal bead arrays. Genome Res 16(3): 383-393.

  • 39. www.illumina.com/pages.ilmn?ID=193 (2007).

  • 40. http://www.illumina.com (2007).

  • 41. Benjamini y, Hochberg y (1995) Controlling the false discovery rate: a practical and powerful approach to multiple testing. R Statist Soc B 57(1): 289-300.

  • 42. Kaufman L, Rousseeuw P J (1990) Finding Groups in Data: An introduction to cluster analysis. Wiley Series in Probability and Mathematical Statistics. New York: John Wiley & Sons, Inc.

  • 43. www.geneimprint.com (2006).

  • 44. Astuti D, Latif F, Wagner K, Gentle D, Cooper W N et al. (2005) Epigenetic alteration at the DLK1-GTL2 imprinted domain in human neoplasia: analysis of neuroblastoma, phaeochromocytoma and Wilms' tumour. Br J Cancer 92(8): 1574-1580

  • 45. Bastepe M, Frohlich L F, Hendy G N, Indridason O S, Josse R G et al. (2003) Autosomal dominant pseudohypoparathyroidism type Ib is associated with a heterozygous microdeletion that likely disrupts a putative imprinting control element of GNAS. J Clin Invest 112(8): 1255-1263.

  • 46. Bastepe M, Frohlich L F, Linglart A, Abu-Zahra H S, Tojo K et al. (2005) Deletion of the NESP55 differentially methylated region causes loss of maternal GNAS imprints and pseudohypoparathyroidism type Ib. Nat Genet 37(1): 25-27.

  • 47. Bell A C, Felsenfeld G (2000) Methylation of a CTCF-dependent boundary controls imprinted expression of the Igf2 gene. Nature 405(6785): 482-485.

  • 48. de la Puente A, Hall J, Wu Y Z, Leone G, Peters J et al. (2002) Structural characterization of Rasgrf1 and a novel linked imprinted locus. Gene 291(1-2): 287-297.

  • 49. Gaston V, Le Bouc Y, Soupre V, Burglen L, Donadieu J et al. (2001) Analysis of the methylation status of the KCNQ10T and H19 genes in leukocyte DNA for the diagnosis and prognosis of Beckwith-Wiedemann syndrome. Eur J Hum Genet 9(6):409-418.

  • 50. Higashimoto K, Soejima H, Saito T, Okumura K, Mukai T (2006) Imprinting disruption of the CDKN1C/KCNQ10T1 domain: the molecular mechanisms causing Beckwith-Wiedemann syndrome and cancer. Cytogenet Genome Res 113(1-4): 306-312.

  • 51. Jie X, Lang C, Jian Q, Chaoqun L, Dehua Y et al. (2007) Androgen activates PEG10 to promote carcinogenesis in hepatic cancer cells. Oncogene.

  • 52. Liu J, Nealon J G, Weinstein L S (2005) Distinct patterns of abnormal GNAS imprinting in familial and sporadic pseudohypoparathyroidism type IB. Hum Mol Genet 14(1): 95-102.

  • 53. Murphy S K, Wylie A A, Jirtle R L (2001) Imprinting of PEG3, the human homologue of a mouse gene involved in nurturing behavior. Genomics 71(1): 110-117.

  • 54. Runte M, Huttenhofer A, Gross S, Kiefmann M, Horsthemke B et al. (2001) The IC-SNURF-SNRPN transcript serves as a host for multiple small nucleolar RNA species and as an antisense RNA for UBE3A. Hum Mol Genet 10(23): 2687-2700.

  • 55. Runte M, Kroisel P M, Gillessen-Kaesbach G, Varon R, Horn D et al. (2004) SNURF-SNRPN and UBE3A transcript levels in patients with Angelman syndrome. Hum Genet 114(6): 553-561.

  • 56. Sutcliffe J S, Nakao M, Christian S, Orstavik K H, Tommerup N et al. (1994) Deletions of a differentially methylated CpG island at the SNRPN gene define a putative imprinting control region. Nat Genet 8(1): 52-58.

  • 57. Suzuki S, Ono R, Narita T, Pask A J, Shaw G et al. (2007) Retrotransposon silencing by DNA methylation can drive mammalian genomic imprinting. PLoS Genet 3(4): e55.

  • 58. Vu T H, Li T, Nguyen D, Nguyen B T, Yao X M et al. (2000) Symmetric and asymmetric DNA methylation in the human IGF2-H19 imprinted region. Genomics 64(2): 132-143.

  • 59. Cui H, Onyango P, Brandenburg S, Wu Y, Hsieh C L et al. (2002) Loss of imprinting in colorectal cancer linked to hypomethylation of H19 and IGF2. Cancer Res 62(22): 6442-6446.

  • 60. Hancock A L, Brown K W, Moorwood K, Moon H, Holmgren C et al. (2007) A CTCF-binding silencer regulates the imprinted genes AWT1 and WT1-AS and exhibits sequential epigenetic defects during Wilms' tumourigenesis. Hum Mol Genet 16(3): 343-354.

  • 61. Kim J D, Hinz A K, Choo J H, Stubbs L, Kim J (2007) YY1 as a controlling factor for the Peg3 and Gnas imprinted domains. Genomics 89(2): 262-269.

  • 62. Lin S P, Youngson N, Takada S, Seitz H, Reik W et al. (2003) Asymmetric regulation of imprinting on the maternal and paternal chromosomes at the DIk1-Gtl2 imprinted cluster on mouse chromosome 12. Nat Genet 35(1): 97-102.

  • 63. Murrell A, Heeson S, Reik W (2004) Interaction between differentially methylated regions partitions the imprinted genes Igf2 and H19 into parent-specific chromatin loops. Nat Genet 36(8): 889-893.

  • 64. Ono R, Kobayashi S, Wagatsuma H, Aisaka K, Kohda T et al. (2001) A retrotransposon-derived gene, PEG10, is a novel imprinted gene located on human chromosome 7q21. Genomics 73(2): 232-237.

  • 65. Sullivan M J, Taniguchi T, Jhee A, Kerr N, Reeve A E (1999) Relaxation of IGF2 imprinting in Wilms tumours associated with specific changes in IGF2 methylation. Oncogene 18(52): 7527-7534.

  • 66. Yun J, Park C W, Lee Y J, Chung J H (2003) Allele-specific methylation at the promoter-associated CpG island of mouse Copg2. Mamm Genome 14(6): 376-382.

  • 67. Reubinoff B E, Pera M F, Fong C Y, Trounson A, Bongso A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol 18 399-404, 2000.


Claims
  • 1. A method for diagnosis or prognosis of testicular or male germ-cell derived cancer, comprising: obtaining a sample of human sperm DNA from a test subject;determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; anddetermining, based on the methylation status of the at least one CpG sequence, diagnosis or prognosis of testicular or male germ-cell derived cancer with respect to the test subject.
  • 2. The method of claim 1, wherein the determined methylation status of the at least one CpG sequence is hypermethylation.
  • 3. The method of claim 1, wherein determining the methylation status of at least one CpG dinucleotide sequence comprises 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.
  • 4. The method of claim 3, wherein treating comprises use of bisulfite treatment of the DNA.
  • 5. The method of claim 1, wherein the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.
  • 6. The method of claim 1, wherein the diagnosis or prognosis is of germ-cell derived testicular cancer.
  • 7. The method of claim 5, comprising determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8 and PLAGL1.
  • 8. The method of claim 7, wherein the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, and PLAGL1 SEQ ID NOS:7 and 19.
  • 9. A method for diagnosis or prognosis of testicular or male germ-cell derived cancer, comprising: obtaining a sample of human sperm DNA from a test subject;determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence from each of a repetitive DNA element sequence group, a maternally imprinted gene sequence group, and a non-imprinted gene sequence group; anddetermining, based on the methylation status of the at least one CpG sequence from each of the groups, diagnosis or prognosis of testicular or male germ-cell derived cancer with respect to the test subject.
  • 10. The method of claim 9, wherein the determined methylation status of the at least one CpG sequence is hypermethylation.
  • 11. The method of claim 9, wherein determining the methylation status of at least one CpG dinucleotide sequence comprises 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.
  • 12. The method of claim 11, wherein treating comprises use of bisulfite treatment of the DNA.
  • 13. The method of claim 9, wherein the at least one gene sequence from a repetitive element group comprises at least one selected from the group consisting of SAT2CHRM1 SEQ ID NOS:9 and 21.
  • 14. The method of claim 9, wherein the at least one gene sequence from a maternally imprinted gene group comprises at least one selected from the group consisting of PLAGL1 SEQ ID NOS:7 and 19, MEST SEQ ID NOS:5 and 17, and DIRAS3 SEQ ID NOS:3 and 15.
  • 15. The method of claim 9, wherein the at least one gene sequence from a non-imprinted gene group comprises at least one selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, SFN SEQ ID NOS:6 and 18, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.
  • 16. A method for screening for agents that cause testicular or male germ-cell derived cancer, comprising: obtaining human ES-cell derived primordial germ cells;contacting the germ cells or descendants thereof, with at least one test agent;culturing the contacted germ cells or the descendants thereof under conditions suitable for germ cell proliferation or development;obtaining a sample of genomic DNA from the contacted cultured germ cells or the descendants thereof;determining, using the genomic DNA of the sample, the methylation status of at least one CpG dinucleotide sequence of at least one gene sequence selected from the group consisting of HRAS, NTF3, MT1A, PAX8, DIRAS3, PLAGL1, SFN, SAT2CHRM1, MEST, RNR1, CYP27B1 and ICAM1; andidentifying, based on the methylation status of the at least one CpG sequence, at least one test agent that causes testicular or male germ-cell derived cancer.
  • 17. The method of claim 16, wherein the determined methylation status of the at least one CpG sequence is hypermethylation.
  • 18. The method of claim 16, wherein determining the methylation status of at least one CpG dinucleotide sequence comprises 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.
  • 19. The method of claim 18, wherein treating comprises use of bisulfite treatment of the DNA.
  • 20. The method of claim 16, wherein the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, DIRAS3 SEQ ID NOS:3 and 15, PLAGL1 SEQ ID NOS:7 and 19, SFN SEQ ID NOS:6 and 18, SAT2CHRM1 SEQ ID NOS:9 and 21, MEST SEQ ID NOS:5 and 17, RNR1 SEQ ID NOS:10 and 22, CYP27B1 SEQ ID NOS:11 and 23 and ICAM1 SEQ ID NOS:12 and 24.
  • 21. The method of claim 20, wherein the at least one gene sequence is selected from the group consisting of HRAS SEQ ID NOS:63 and 20, NTF3 SEQ ID NOS:2 and 14, MT1A SEQ ID NOS:4 and 16, PAX8 SEQ ID NOS:1 and 13, and PLAGL1 SEQ ID NOS:7 and 19.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of priority to U.S. Provisional Patent Application Ser. No. 61/110,890, filed 3 Nov. 2008 and entitled COMPOSITIONS AND METHODS FOR DIAGNOSIS OR PROGNOSIS OF TESTICULAR CANCER, which is incorporated herein by reference in its entirety.

FEDERAL FUNDING ACKNOWLEDGEMENT

This work was at least in part supported by the Southern California Environmental Health Sciences Center Grant No. 5P30ES007048, funded by the National Institute of Environmental Health Sciences. The United States government therefore has certain rights in the invention.

Provisional Applications (1)
Number Date Country
61110890 Nov 2008 US