X. D. Zhao, Whole-genome mapping of histone H3 Lys4 and 27 trimethylations reveals distinct genomic compartments in human embryonic stem cells, Cell stem cell, vol.1, pp.286-298, 2007.

A. Barski, High-resolution profiling of histone methylations in the human genome, Cell, vol.129, pp.823-837, 2007.

T. S. Mikkelsen, Genome-wide maps of chromatin state in pluripotent and lineage-committed cells, Nature, vol.448, pp.553-560, 2007.

B. J. Lesch, G. A. Dokshin, R. A. Young, J. R. Mccarrey, and D. C. Page, A set of genes critical to development is epigenetically poised in mouse germ cells from fetal stages through completion of meiosis, Proceedings of the National Academy of Sciences of the United States of America, vol.110, pp.16061-16066, 2013.

U. Brykczynska, Repressive and active histone methylation mark distinct promoters in human and mouse spermatozoa, Nature structural & molecular biology, vol.17, pp.679-687, 2010.

J. Bao and M. T. Bedford, Epigenetic regulation of the histone-to-protamine transition during spermiogenesis, Reproduction, vol.151, pp.55-70, 2016.

K. Siklenka, Disruption of histone methylation in developing sperm impairs offspring health transgenerationally, Science, vol.350, 2006.

B. Samans, Uniformity of nucleosome preservation pattern in Mammalian sperm and its connection to repetitive DNA elements, Developmental cell, vol.30, pp.23-35, 2014.

H. Zheng, Resetting Epigenetic Memory by Reprogramming of Histone Modifications in Mammals, Mol Cell, vol.63, pp.1066-1079, 2016.

B. Zhang, Allelic reprogramming of the histone modification H3K4me3 in early mammalian development, Nature, vol.537, pp.553-557, 2016.

M. D. Anway, A. S. Cupp, M. Uzumcu, and M. K. Skinner, Epigenetic transgenerational actions of endocrine disruptors and male fertility, Science, vol.308, pp.1466-1469, 2005.

M. D. Anway, S. S. Rekow, and M. K. Skinner, Comparative anti-androgenic actions of vinclozolin and flutamide on transgenerational adult onset disease and spermatogenesis, Reproductive toxicology, vol.26, pp.100-106, 2008.

R. Tracey, M. Manikkam, C. Guerrero-bosagna, and M. K. Skinner, Hydrocarbons (jet fuel JP-8) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations, Reproductive toxicology, vol.36, pp.104-116, 2013.

C. Hao, Exposure to the widely used herbicide atrazine results in deregulation of global tissue-specific RNA transcription in the third generation and is associated with a global decrease of histone trimethylation in mice, Nucleic acids research, vol.44, pp.9784-9802, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01477213

M. Trerotola, V. Relli, P. Simeone, and S. Alberti, Epigenetic inheritance and the missing heritability, Human genomics, vol.9, 2015.

E. Heard and R. A. Martienssen, Transgenerational epigenetic inheritance: myths and mechanisms, ScientiFic RePoRTS |, vol.8, pp.95-109, 2014.

M. D. Reuber, Carcinomas of the liver in rats ingesting kepone, Neoplasma, vol.26, pp.231-235, 1979.

M. D. Reuber, The carcinogenicity kepone, Journal of environmental pathology and toxicology, vol.2, pp.671-686, 1979.

A. E. Sirica, Evaluation of chlordecone in a two-stage model of hepatocarcinogenesis: a significant sex difference in the hepatocellular carcinoma incidence, Carcinogenesis, vol.10, pp.1047-1054, 1989.

O. Gandolfi, D. L. Cheney, J. S. Hong, and E. Costa, On the neurotoxicity of chlordecone: a role for gamma-aminobutyric acid and serotonin, Brain research, vol.303, pp.117-123, 1984.

G. G. Kuiper, Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta, Endocrinology, vol.139, pp.4252-4263, 1998.

G. Lemaire, W. Mnif, P. Mauvais, P. Balaguer, and R. Rahmani, Activation of alpha-and beta-estrogen receptors by persistent pesticides in reporter cell lines, Life sciences, vol.79, pp.1160-1169, 2006.

S. J. Ellem and G. P. Risbridger, The dual, opposing roles of estrogen in the prostate, Annals of the New York Academy of Sciences, vol.1155, pp.174-186, 2009.

J. R. Taylor, J. B. Selhorst, S. A. Houff, and A. J. Martinez, Chlordecone intoxication in man. I. Clinical observations, Neurology, vol.28, pp.626-630, 1978.

S. B. Cannon, Epidemic kepone poisoning in chemical workers, American journal of epidemiology, vol.107, pp.529-537, 1978.

L. Multigner, Chlordecone exposure and risk of prostate cancer, Journal of clinical oncology: official journal of the American Society of Clinical Oncology, vol.28, pp.3457-3462, 2010.
DOI : 10.1200/jco.2009.27.2153

URL : https://hal.archives-ouvertes.fr/inserm-00518099

O. Faroon, S. Kueberuwa, L. Smith, and C. Derosa, ATSDR evaluation of health effects of chemicals. II. Mirex and chlordecone: health effects, toxicokinetics, human exposure, and environmental fate, Toxicology and industrial health, vol.11, pp.1-203, 1995.

P. Kadhel, Chlordecone exposure, length of gestation, and risk of preterm birth, American journal of epidemiology, vol.179, pp.536-544, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01010740

D. Herve, Prenatal exposure to chlordecone, gestational weight gain, and birth weight in a Guadeloupean birth cohort, Environmental research, vol.151, pp.436-444, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01367154

N. Costet, Perinatal exposure to chlordecone and infant growth, Environmental research, vol.142, pp.123-134, 2015.
DOI : 10.1016/j.envres.2015.06.023

URL : https://hal.archives-ouvertes.fr/hal-01169790

O. Boucher, Exposure to an organochlorine pesticide (chlordecone) and development of 18-month-old infants, Neurotoxicology, vol.35, pp.162-168, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00875907

K. Hayashi, S. M. De-sousa-lopes, and M. A. Surani, Germ cell specification in mice, Science, vol.316, pp.394-396, 2007.

Y. Ohinata, Blimp1 is a critical determinant of the germ cell lineage in mice, Nature, vol.436, pp.207-213, 2005.

A. , Toxicological profile for Mirex and Chlordecone, Public Health Service, 1995.

M. K. Skinner, Environmentally induced transgenerational epigenetic reprogramming of primordial germ cells and the subsequent germ line, PloS one, vol.8, 2013.

P. Hajkova, Epigenetic reprogramming in mouse primordial germ cells, Mechanisms of development, vol.117, pp.15-23, 2002.
DOI : 10.1016/s0925-4773(02)00181-8

URL : https://doi.org/10.1016/s0925-4773(02)00181-8

A. Schuster, M. K. Skinner, and W. Yan, Ancestral vinclozolin exposure alters the epigenetic transgenerational inheritance of sperm small noncoding RNAs, Environmental epigenetics, vol.2, 2016.

D. Beck, I. Sadler-riggleman, and M. K. Skinner, Generational comparisons (F1 versus F3) of vinclozolin induced epigenetic transgenerational inheritance of sperm differential DNA methylation regions (epimutations) using MeDIP-Seq, Environmental epigenetics, vol.3, 2017.

L. A. Annab, Differential responses to retinoic acid and endocrine disruptor compounds of subpopulations within human embryonic stem cell lines. Differentiation; research in biological diversity, vol.84, pp.330-343, 2012.

K. A. Henderson and S. Keeney, Tying synaptonemal complex initiation to the formation and programmed repair of DNA doublestrand breaks, Proceedings of the National Academy of Sciences of the United States of America, vol.101, pp.4519-4524, 2004.

Z. Li, E. I. Golub, R. Gupta, and C. M. Radding, Recombination activities of HsDmc1 protein, the meiotic human homolog of RecA protein, Proceedings of the National Academy of Sciences of the United States of America, vol.94, pp.11221-11226, 1997.

T. Kouzarides, Chromatin modifications and their function, Cell, vol.128, pp.693-705, 2007.

M. Vermeulen, Quantitative interaction proteomics and genome-wide profiling of epigenetic histone marks and their readers, Cell, vol.142, pp.967-980, 2010.

D. K. Goode, Dynamic Gene Regulatory Networks Drive Hematopoietic Specification and Differentiation, Developmental cell, vol.36, pp.572-587, 2016.

C. E. Grant, T. L. Bailey, and W. S. Noble, FIMO: scanning for occurrences of a given motif, Bioinformatics, vol.27, pp.1017-1018, 2011.

R. C. Mcleay and T. L. Bailey, Motif Enrichment Analysis: a unified framework and an evaluation on ChIP data, BMC bioinformatics, vol.11, 2010.

X. Dong and Z. Weng, The correlation between histone modifications and gene expression, Epigenomics, vol.5, pp.113-116, 2013.

S. Xiao, Comparative epigenomic annotation of regulatory DNA, Cell, vol.149, pp.1381-1392, 2012.

Y. H. Woo and W. H. Li, Evolutionary conservation of histone modifications in mammals, Molecular biology and evolution, vol.29, pp.1757-1767, 2012.

N. S. Christophersen and K. Helin, Epigenetic control of embryonic stem cell fate, The Journal of experimental medicine, vol.207, pp.2287-2295, 2010.

D. Zeineddine, A. A. Hammoud, M. Mortada, and H. Boeuf, The Oct4 protein: more than a magic stemness marker, American journal of stem cells, vol.3, pp.74-82, 2014.

M. Pesce and H. R. Scholer, Oct-4: gatekeeper in the beginnings of mammalian development, Stem cells, vol.19, pp.271-278, 2001.

J. Kehler, Oct4 is required for primordial germ cell survival, EMBO reports, vol.5, pp.1078-1083, 2004.

T. Akagi, Identification of Zfp-57 as a downstream molecule of STAT3 and Oct-3/4 in embryonic stem cells, Biochemical and biophysical research communications, vol.331, pp.23-30, 2005.

D. J. Mackay, Hypomethylation of multiple imprinted loci in individuals with transient neonatal diabetes is associated with mutations in ZFP57, Nature genetics, vol.40, pp.949-951, 2008.

N. Takahashi, ZFP57 and the Targeted Maintenance of Postfertilization Genomic Imprints, Cold Spring Harbor symposia on quantitative biology, vol.80, pp.177-187, 2015.

, ScientiFic RePoRTS |, vol.8, 2018.

J. Kent, S. C. Wheatley, J. E. Andrews, A. H. Sinclair, and P. Koopman, A male-specific role for SOX9 in vertebrate sex determination, Development, vol.122, pp.2813-2822, 1996.

T. X. Garcia and M. C. Hofmann, NOTCH signaling in Sertoli cells regulates gonocyte fate, Cell cycle, vol.12, pp.2538-2545, 2013.

A. Sfeir and T. De-lange, Removal of shelterin reveals the telomere end-protection problem, Science, vol.336, pp.593-597, 2012.

R. E. Linder, Spermotoxicity and tissue accumulation of chlordecone (Kepone) in male rats, Journal of toxicology and environmental health, vol.12, pp.183-192, 1983.

J. Ding, FDA-approved medications that impair human spermatogenesis, Oncotarget, vol.8, pp.10714-10725, 2017.
DOI : 10.18632/oncotarget.12956

URL : http://www.oncotarget.com/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=12956&path%5B%5D=41046

X. Li, A maternal-zygotic effect gene, Zfp57, maintains both maternal and paternal imprints, Developmental cell, vol.15, pp.547-557, 2008.
DOI : 10.1016/j.devcel.2008.08.014

URL : https://doi.org/10.1016/j.devcel.2008.08.014

R. Strogantsev and A. C. Ferguson-smith, Proteins involved in establishment and maintenance of imprinted methylation marks, Briefings in functional genomics, vol.11, pp.227-239, 2012.

V. Riso, ZFP57 maintains the parent-of-origin-specific expression of the imprinted genes and differentially affects nonimprinted targets in mouse embryonic stem cells, Nucleic acids research, vol.44, pp.8165-8178, 2016.

M. Gloerich, J. M. Bianchini, K. A. Siemers, D. J. Cohen, and W. J. Nelson, Cell division orientation is coupled to cell-cell adhesion by the E-cadherin/LGN complex, Nature communications, vol.8, 2017.
DOI : 10.1038/ncomms13996

URL : http://www.nature.com/articles/ncomms13996.pdf

A. Goudarzi, Dynamic Competing Histone H4 K5K8 Acetylation and Butyrylation Are Hallmarks of Highly Active Gene Promoters, Molecular cell, vol.62, pp.169-180, 2016.
DOI : 10.1016/j.molcel.2016.03.014

URL : https://hal.archives-ouvertes.fr/hal-01341099

V. Bourdeau, Genome-wide identification of high-affinity estrogen response elements in human and mouse, Molecular endocrinology, vol.18, pp.1411-1427, 2004.

J. S. Carroll, Genome-wide analysis of estrogen receptor binding sites, Nature genetics, vol.38, pp.1289-1297, 2006.
DOI : 10.1038/ng1901

S. R. Joshi, R. B. Ghattamaneni, and W. M. Scovell, Expanding the paradigm for estrogen receptor binding and transcriptional activation, Molecular endocrinology, vol.25, pp.980-994, 2011.
DOI : 10.1210/me.2010-0302

URL : https://academic.oup.com/mend/article-pdf/25/6/980/8931750/mend0980.pdf

L. Magnani and M. Lupien, Chromatin and epigenetic determinants of estrogen receptor alpha (ESR1) signaling, Molecular and cellular endocrinology, vol.382, pp.633-641, 2014.
DOI : 10.1016/j.mce.2013.04.026

H. J. Li, NKX6.1 functions as a metastatic suppressor through epigenetic regulation of the epithelial-mesenchymal transition, Oncogene, vol.35, pp.2266-2278, 2016.

B. Xu, M. Zhou, W. Qiu, J. Ye, and Q. Feng, CCR7 mediates human breast cancer cell invasion, migration by inducing epithelialmesenchymal transition and suppressing apoptosis through AKT pathway, Cancer medicine, vol.6, pp.1062-1071, 2017.
DOI : 10.1002/cam4.1039

URL : http://onlinelibrary.wiley.com/doi/10.1002/cam4.1039/pdf

M. P. Ribeiro, A. E. Santos, and J. B. Custodio, Interplay between estrogen and retinoid signaling in breast cancer-current and future perspectives, Cancer letters, vol.353, pp.17-24, 2014.
DOI : 10.1016/j.canlet.2014.07.009

A. Zsarnovszky, Thyroid hormone-and estrogen receptor interactions with natural ligands and endocrine disruptors in the cerebellum, Frontiers in neuroendocrinology, 2017.
DOI : 10.1016/j.yfrne.2017.10.001

X. Dong, Modeling gene expression using chromatin features in various cellular contexts, Genome biology, vol.13, 2012.
DOI : 10.1186/gb-2012-13-9-r53

URL : https://genomebiology.biomedcentral.com/track/pdf/10.1186/gb-2012-13-9-r53?site=genomebiology.biomedcentral.com

X. Zhang, Systematic identification and characterization of long non-coding RNAs in mouse mature sperm, PloS one, vol.12, 2017.

P. S. Larson, J. L. Egle, . Jr, G. R. Hennigar, R. W. Lane et al., Acute, subchronic, and chronic toxicity of chlordecone, Toxicology and applied pharmacology, vol.48, pp.29-41, 1979.
DOI : 10.1016/s0041-008x(79)80005-8

V. Vallet-erdtmann, The testicular form of hormone-sensitive lipase HSLtes confers rescue of male infertility in HSL-deficient mice, The Journal of biological chemistry, vol.279, pp.42875-42880, 2004.

A. H. Peters, A. W. Plug, M. J. Van-vugt, and P. De-boer, Chromosome research: an international journal on the molecular, supramolecular and evolutionary aspects of chromosome biology, vol.5, pp.66-68, 1997.

D. Kim, TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions, Genome biology, vol.14, 2013.

C. Trapnell, Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks, Nature protocols, vol.7, pp.562-578, 2012.
DOI : 10.1038/nprot.2012.016

URL : https://authors.library.caltech.edu/74752/3/nihms-366741.pdf

M. E. Ritchie, limma powers differential expression analyses for RNA-sequencing and microarray studies, Nucleic acids research, vol.43, 2015.
DOI : 10.1093/nar/gkv007

URL : https://academic.oup.com/nar/article-pdf/43/7/e47/7207289/gkv007.pdf

H. Thorvaldsdottir, J. T. Robinson, and J. P. Mesirov, Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration, Briefings in bioinformatics, vol.14, pp.178-192, 2013.
DOI : 10.1093/bib/bbs017

URL : https://academic.oup.com/bib/article-pdf/14/2/178/546734/bbs017.pdf

A. Gely-pernot, The epigenetic processes of meiosis in male mice are broadly affected by the widely used herbicide atrazine, BMC genomics, vol.16, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01225493

A. Diaz, A. Nellore, and J. S. Song, CHANCE: comprehensive software for quality control and validation of ChIP-seq data, Genome biology, vol.13, 2012.
DOI : 10.1186/gb-2012-13-10-r98

URL : https://genomebiology.biomedcentral.com/track/pdf/10.1186/gb-2012-13-10-r98?site=genomebiology.biomedcentral.com

J. Feng, T. Liu, B. Qin, Y. Zhang, and X. S. Liu, Identifying ChIP-seq enrichment using MACS, Nature protocols, vol.7, pp.1728-1740, 2012.
DOI : 10.1038/nprot.2012.101

URL : http://europepmc.org/articles/pmc3868217?pdf=render

H. Shin, T. Liu, A. K. Manrai, and X. S. Liu, CEAS: cis-regulatory element annotation system, Bioinformatics, vol.25, pp.2605-2606, 2009.

W. Huang-da, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nat Protoc, vol.4, pp.44-57, 2009.

C. Y. Mclean, GREAT improves functional interpretation of cis-regulatory regions, Nat Biotechnol, vol.28, pp.495-501, 2010.

E. Y. Chen, Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool, BMC bioinformatics, vol.14, 2013.
DOI : 10.1186/1471-2105-14-128

URL : https://bmcbioinformatics.biomedcentral.com/track/pdf/10.1186/1471-2105-14-128

M. V. Kuleshov, Enrichr: a comprehensive gene set enrichment analysis web server 2016 update, Nucleic acids research, vol.44, pp.90-97, 2016.
DOI : 10.1093/nar/gkw377

URL : https://academic.oup.com/nar/article-pdf/44/W1/W90/18788036/gkw377.pdf

A. Lachmann, ChEA: transcription factor regulation inferred from integrating genome-wide ChIP-X experiments, Bioinformatics, vol.26, pp.2438-2444, 2010.
DOI : 10.1093/bioinformatics/btq466

URL : https://academic.oup.com/bioinformatics/article-pdf/26/19/2438/16896569/btq466.pdf