Advertisement

Generations affected by histone changes

Parent and even grandparent environmental exposure can transmit adverse health effects to offspring. The mechanism of transmission is unclear, but some studies have implicated variations in DNA methylation. In a mouse model, Siklenka et al. found that alterations in histone methylation during sperm formation in one generation leads to reduced survival and developmental abnormalities in three subsequent generations (see the Perspective by McCarrey). Although changes in DNA methylation were not observed, altered sperm RNA content and abnormal gene expression in offspring were measured. Thus, chromatin may act as a mediator of molecular memory in transgenerational inheritance.
Science, this issue p. 10.1126/science.aab2006; see also p. 634

Structured Abstract

INTRODUCTION

Despite the father transmitting half of the heritable information to the embryo, the focus of preconception health has been the mother. Paternal effects have been linked to complex diseases such as cancer, diabetes, and obesity. These diseases are increasing in prevalence at rates that cannot be explained by genetics alone and highlight the potential for disease transmission via nongenetic inheritance, through epigenetic mechanisms. Epigenetic mechanisms include DNA methylation, posttranslational modifications of histones, and noncoding RNA. Studies in humans and animals suggest that epigenetic mechanisms may serve in the transmission of environmentally induced phenotypic traits from the father to the offspring. Such traits have been associated with altered gene expression and tissue function in first and second offspring generations, a phenomenon known as intergenerational or transgenerational inheritance, respectively. The mechanisms underlying such paternal epigenetic transmission are unclear.

RATIONALE

Sperm formation involves rapid cell division and distinctive transcription programs, resulting in a motile cell with highly condensed chromatin. Within the highly compacted sperm nucleus, few histones are retained in a manner that suggests an influential role in development. Despite being the major focus of studies in epigenetic inheritance, the role of DNA methylation in paternal epigenetic inheritance is unresolved, as only minor changes in DNA methylation in sperm at CpG-enriched regions have been associated with transmission of environmentally induced traits. Instead, there may be a combination of molecular mechanisms underlying paternal transgenerational epigenetic inheritance involving changes in histone states and/or RNA in sperm. The function of sperm histones and their modifications in embryonic development, offspring health, and epigenetic inheritance is unknown. By overexpressing the human KDM1A histone lysine 4 demethylase during mouse spermatogenesis, we generated a mouse model producing spermatozoa with reduced H3K4me2 within the CpG islands of genes implicated in development, and we studied the development and fitness of the offspring.

RESULTS

Male transgenic offspring were bred with C57BL/6 females, generating the experimental heterozygous transgenic (TG) and nontransgenic (nonTG) brothers. Each generation from TG and nonTG animals (F1 to F3 in our transgenerational studies) was bred with C57BL/6 females, and the offspring (pups from generations F1 to F4) were analyzed for intergenerational and transgenerational effects. We found that KDM1A overexpression in one generation severely impaired development and survivability of offspring. These defects lasted for two subsequent generations in the absence of KDM1A germline expression. We characterized histone and DNA methylation states in the sperm of TG and nonTG sires. Overexpression of KDM1A was associated with a specific loss of H3K4me2 at more than 2300 genes, including many developmental regulatory genes. Unlike in other examples of paternal transgenerational inheritance, we observed no changes in sperm DNA methylation associated with primarily CpG-enriched regions. Instead, we measured robust and analogous changes in sperm RNA content of TG and nonTG descendants, as well as in their offspring, at the two-cell stage. These changes in expression and the phenotypic abnormalities observed in offspring correlated with altered histone methylation levels at genes in sperm. This study demonstrates that KDM1A activity during sperm development has major developmental consequences for offspring and implicates histone methylation and sperm RNA as potential mediators of transgenerational inheritance. Our data emphasize the complexity of transgenerational epigenetic inheritance likely involving multiple molecular factors, including the establishment of chromatin states in spermatogenesis and sperm-borne RNA.

CONCLUSION

Correct histone methylation during spermatogenesis is critical for offspring development and survival over multiple generations. These findings demonstrate the potential of histone methylation as a molecular mechanism underlying paternal epigenetic inheritance. Its modification by environmental influences may alter embryo development and complex disease transmission across generations. An essential next step is to establish functional links between environmental exposures, the composition of the sperm epigenome, and consequent altered gene expression and metabolic processes in offspring. Considering the mounting evidence, it may soon be reasonable to suggest that future fathers protect their sperm epigenome.
Disruption of histone methylation in developing sperm by exposure to the KDM1A transgene in one generation severely impaired development and survivability of offspring.
These defects were transgenerational and occurred in nonTG descendants in the absence of KDM1A germline expression. Developmental defects in offspring and embryos were associated with altered RNA expression in sperm and embryos.

Abstract

A father’s lifetime experiences can be transmitted to his offspring to affect health and development. However, the mechanisms underlying paternal epigenetic transmission are unclear. Unlike in somatic cells, there are few nucleosomes in sperm, and their function in epigenetic inheritance is unknown. We generated transgenic mice in which overexpression of the histone H3 lysine 4 (H3K4) demethylase KDM1A (also known as LSD1) during spermatogenesis reduced H3K4 dimethylation in sperm. KDM1A overexpression in one generation severely impaired development and survivability of offspring. These defects persisted transgenerationally in the absence of KDM1A germline expression and were associated with altered RNA profiles in sperm and offspring. We show that epigenetic inheritance of aberrant development can be initiated by histone demethylase activity in developing sperm, without changes to DNA methylation at CpG-rich regions.

Get full access to this article

View all available purchase options and get full access to this article.

Supplementary Material

Summary

Figs. S1 to S9
Tables S1 to S7
Reference (65)

Resources

File (aab2006_table_s2.xlsx)
File (aab2006_table_s3.xlsx)
File (aab2006_table_s4.xlsx)
File (aab2006_table_s5.xlsx)
File (aab2006_table_s6.xlsx)
File (aab2006_table_s7.xlsx)
File (siklenka.sm.pdf)

References and Notes

1
Lobo I., Zhaurova K., Birth defects: causes and statistics. Nat. Educ. 1, 18 (2008).
2
T. J. Mathews, M. F. MacDorman, “Infant mortality statistics from the 2005 period linked birth/infant death data set” (National Vital Statistics Reports, National Center for Health Statistics, Hyattsville, MD, 2008), vol. 57.
3
Anway M. D., Cupp A. S., Uzumcu M., Skinner M. K., Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466–1469 (2005).
4
Pembrey M. E., Bygren L. O., Kaati G., Edvinsson S., Northstone K., Sjöström M., Golding J.ALSPAC Study Team, Sex-specific, male-line transgenerational responses in humans. Eur. J. Hum. Genet. 14, 159–166 (2006).
5
Carone B. R., Fauquier L., Habib N., Shea J. M., Hart C. E., Li R., Bock C., Li C., Gu H., Zamore P. D., Meissner A., Weng Z., Hofmann H. A., Friedman N., Rando O. J., Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 143, 1084–1096 (2010).
6
Ng S. F., Lin R. C., Laybutt D. R., Barres R., Owens J. A., Morris M. J., Chronic high-fat diet in fathers programs β-cell dysfunction in female rat offspring. Nature 467, 963–966 (2010).
7
Lambrot R., Xu C., Saint-Phar S., Chountalos G., Cohen T., Paquet M., Suderman M., Hallett M., Kimmins S., Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat. Commun. 4, 2889 (2013).
8
Dias B. G., Ressler K. J., Parental olfactory experience influences behavior and neural structure in subsequent generations. Nat. Neurosci. 17, 89–96 (2014).
9
Skinner M. K., Guerrero-Bosagna C., Role of CpG deserts in the epigenetic transgenerational inheritance of differential DNA methylation regions. BMC Genomics 15, 692 (2014).
10
Zeybel M., Hardy T., Wong Y. K., Mathers J. C., Fox C. R., Gackowska A., Oakley F., Burt A. D., Wilson C. L., Anstee Q. M., Barter M. J., Masson S., Elsharkawy A. M., Mann D. A., Mann J., Multigenerational epigenetic adaptation of the hepatic wound-healing response. Nat. Med. 18, 1369–1377 (2012).
11
Radford E. J., Ito M., Shi H., Corish J. A., Yamazawa K., Isganaitis E., Seisenberger S., Hore T. A., Reik W., Erkek S., Peters A. H., Patti M. E., Ferguson-Smith A. C., In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science 345, 1255903 (2014).
12
Bestor T. H., Bourc’his D., Transposon silencing and imprint establishment in mammalian germ cells. Cold Spring Harb. Symp. Quant. Biol. 69, 381–387 (2004).
13
Smallwood S. A., Tomizawa S., Krueger F., Ruf N., Carli N., Segonds-Pichon A., Sato S., Hata K., Andrews S. R., Kelsey G., Dynamic CpG island methylation landscape in oocytes and preimplantation embryos. Nat. Genet. 43, 811–814 (2011).
14
Kobayashi H., Sakurai T., Imai M., Takahashi N., Fukuda A., Yayoi O., Sato S., Nakabayashi K., Hata K., Sotomaru Y., Suzuki Y., Kono T., Contribution of intragenic DNA methylation in mouse gametic DNA methylomes to establish oocyte-specific heritable marks. PLOS Genet. 8, e1002440 (2012).
15
Smith Z. D., Chan M. M., Mikkelsen T. S., Gu H., Gnirke A., Regev A., Meissner A., A unique regulatory phase of DNA methylation in the early mammalian embryo. Nature 484, 339–344 (2012).
16
Brykczynska U., Hisano M., Erkek S., Ramos L., Oakeley E. J., Roloff T. C., Beisel C., Schübeler D., Stadler M. B., Peters A. H., Repressive and active histone methylation mark distinct promoters in human and mouse spermatozoa. Nat. Struct. Mol. Biol. 17, 679–687 (2010).
17
Erkek S., Hisano M., Liang C.-Y., Gill M., Murr R., Dieker J., Schübeler D., van der Vlag J., Stadler M. B., Peters A. H F. M., Molecular determinants of nucleosome retention at CpG-rich sequences in mouse spermatozoa. Nat. Struct. Mol. Biol. 20, 868–875 (2013).
18
Hammoud S. S., Nix D. A., Zhang H., Purwar J., Carrell D. T., Cairns B. R., Distinctive chromatin in human sperm packages genes for embryo development. Nature 460, 473–478 (2009).
19
Sendler E., Johnson G. D., Mao S., Goodrich R. J., Diamond M. P., Hauser R., Krawetz S. A., Stability, delivery and functions of human sperm RNAs at fertilization. Nucleic Acids Res. 41, 4104–4117 (2013).
20
Balhorn R., The protamine family of sperm nuclear proteins. Genome Biol. 8, 227 (2007).
21
Carone B. R., Hung J. H., Hainer S. J., Chou M. T., Carone D. M., Weng Z., Fazzio T. G., Rando O. J., High-resolution mapping of chromatin packaging in mouse embryonic stem cells and sperm. Dev. Cell 30, 11–22 (2014).
22
Saitou M., Kurimoto K., Paternal nucleosomes: Are they retained in developmental promoters or gene deserts? Dev. Cell 30, 6–8 (2014).
23
Vavouri T., Lehner B., Chromatin organization in sperm may be the major functional consequence of base composition variation in the human genome. PLOS Genet. 7, e1002036 (2011).
24
Katz D. J., Edwards T. M., Reinke V., Kelly W. G., A C. elegans LSD1 demethylase contributes to germline immortality by reprogramming epigenetic memory. Cell 137, 308–320 (2009).
25
Shi Y., Lan F., Matson C., Mulligan P., Whetstine J. R., Cole P. A., Casero R. A., Shi Y., Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953 (2004).
26
Guerrero-Bosagna C., Covert T. R., Haque M. M., Settles M., Nilsson E. E., Anway M. D., Skinner M. K., Epigenetic transgenerational inheritance of vinclozolin induced mouse adult onset disease and associated sperm epigenome biomarkers. Reprod. Toxicol. 34, 694–707 (2012).
27
Wei Y., Yang C. R., Wei Y. P., Zhao Z. A., Hou Y., Schatten H., Sun Q. Y., Paternally induced transgenerational inheritance of susceptibility to diabetes in mammals. Proc. Natl. Acad. Sci. U.S.A. 111, 1873–1878 (2014).
28
Furuchi T., Masuko K., Nishimune Y., Obinata M., Matsui Y., Inhibition of testicular germ cell apoptosis and differentiation in mice misexpressing Bcl-2 in spermatogonia. Development 122, 1703–1709 (1996).
29
Blanco-Rodríguez J., γH2AX marks the main events of the spermatogenic process. Microsc. Res. Tech. 72, 823–832 (2009).
30
Cho C., Willis W. D., Goulding E. H., Jung-Ha H., Choi Y. C., Hecht N. B., Eddy E. M., Haploinsufficiency of protamine-1 or -2 causes infertility in mice. Nat. Genet. 28, 82–86 (2001).
31
Braun R. E., Behringer R. R., Peschon J. J., Brinster R. L., Palmiter R. D., Genetically haploid spermatids are phenotypically diploid. Nature 337, 373–376 (1989).
32
Hisano M., Erkek S., Dessus-Babus S., Ramos L., Stadler M. B., Peters A. H., Genome-wide chromatin analysis in mature mouse and human spermatozoa. Nat. Protoc. 8, 2449–2470 (2013).
33
Zhang J., Bonasio R., Strino F., Kluger Y., Holloway J. K., Modzelewski A. J., Cohen P. E., Reinberg D., SFMBT1 functions with LSD1 to regulate expression of canonical histone genes and chromatin-related factors. Genes Dev. 27, 749–766 (2013).
34
Hashimoto N., Kido Y., Uchida T., Asahara S., Shigeyama Y., Matsuda T., Takeda A., Tsuchihashi D., Nishizawa A., Ogawa W., Fujimoto Y., Okamura H., Arden K. C., Herrera P. L., Noda T., Kasuga M., Ablation of PDK1 in pancreatic β cells induces diabetes as a result of loss of β cell mass. Nat. Genet. 38, 589–593 (2006).
35
Collins B. J., Deak M., Murray-Tait V., Storey K. G., Alessi D. R., In vivo role of the phosphate groove of PDK1 defined by knockin mutation. J. Cell Sci. 118, 5023–5034 (2005).
36
Lawlor M. A., Mora A., Ashby P. R., Williams M. R., Murray-Tait V., Malone L., Prescott A. R., Lucocq J. M., Alessi D. R., Essential role of PDK1 in regulating cell size and development in mice. EMBO J. 21, 3728–3738 (2002).
37
Courel M., Friesenhahn L., Lees J. A., E2f6 and Bmi1 cooperate in axial skeletal development. Dev. Dyn. 237, 1232–1242 (2008).
38
Blum M., Gaunt S. J., Cho K. W., Steinbeisser H., Blumberg B., Bittner D., De Robertis E. M., Gastrulation in the mouse: The role of the homeobox gene goosecoid. Cell 69, 1097–1106 (1992).
39
Metzger E., Wissmann M., Yin N., Müller J. M., Schneider R., Peters A. H., Günther T., Buettner R., Schüle R., LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 437, 436–439 (2005).
40
Wissmann M., Yin N., Müller J. M., Greschik H., Fodor B. D., Jenuwein T., Vogler C., Schneider R., Günther T., Buettner R., Metzger E., Schüle R., Cooperative demethylation by JMJD2C and LSD1 promotes androgen receptor-dependent gene expression. Nat. Cell Biol. 9, 347–353 (2007).
41
Lienert F., Mohn F., Tiwari V. K., Baubec T., Roloff T. C., Gaidatzis D., Stadler M. B., Schübeler D., Genomic prevalence of heterochromatic H3K9me2 and transcription do not discriminate pluripotent from terminally differentiated cells. PLOS Genet. 7, e1002090 (2011).
42
Youngson N. A., Whitelaw E., Transgenerational epigenetic effects. Annu. Rev. Genomics Hum. Genet. 9, 233–257 (2008).
43
Lalancette C., Miller D., Li Y., Krawetz S. A., Paternal contributions: New functional insights for spermatozoal RNA. J. Cell. Biochem. 104, 1570–1579 (2008).
44
Russell L. B., Hunsicker P. R., Russell W. L., Comparison of the genetic effects of equimolar doses of ENU and MNU: While the chemicals differ dramatically in their mutagenicity in stem-cell spermatogonia, both elicit very high mutation rates in differentiating spermatogonia. Mutat. Res. 616, 181–195 (2007).
45
Burr K. L., van Duyn-Goedhart A., Hickenbotham P., Monger K., van Buul P. P., Dubrova Y. E., The effects of MSH2 deficiency on spontaneous and radiation-induced mutation rates in the mouse germline. Mutat. Res. 617, 147–151 (2007).
46
Barber R. C., Hardwick R. J., Shanks M. E., Glen C. D., Mughal S. K., Voutounou M., Dubrova Y. E., The effects of in utero irradiation on mutation induction and transgenerational instability in mice. Mutat. Res. 664, 6–12 (2009).
47
Ihara M., Meyer-Ficca M. L., Leu N. A., Rao S., Li F., Gregory B. D., Zalenskaya I. A., Schultz R. M., Meyer R. G., Paternal poly (ADP-ribose) metabolism modulates retention of inheritable sperm histones and early embryonic gene expression. PLOS Genet. 10, e1004317 (2014).
48
Carrell D. T., Elucidating the genetics of male infertility: Understanding transcriptional and translational regulatory networks involved in spermatogenesis. Int. J. Androl. 31, 455–456 (2008).
49
Chong S., Vickaryous N., Ashe A., Zamudio N., Youngson N., Hemley S., Stopka T., Skoultchi A., Matthews J., Scott H. S., de Kretser D., O’Bryan M., Blewitt M., Whitelaw E., Modifiers of epigenetic reprogramming show paternal effects in the mouse. Nat. Genet. 39, 614–622 (2007).
50
Rechavi O., Minevich G., Hobert O., Transgenerational inheritance of an acquired small RNA-based antiviral response in C. elegans. Cell 147, 1248–1256 (2011).
51
Sabin L. R., Delás M. J., Hannon G. J., Dogma derailed: The many influences of RNA on the genome. Mol. Cell 49, 783–794 (2013).
52
Ulitsky I., Bartel D. P., lincRNAs: Genomics, evolution, and mechanisms. Cell 154, 26–46 (2013).
53
Borges F., Martienssen R. A., Establishing epigenetic variation during genome reprogramming. RNA Biol. 10, 490–494 (2013).
54
Sauvageau M., Goff L. A., Lodato S., Bonev B., Groff A. F., Gerhardinger C., Sanchez-Gomez D. B., Hacisuleyman E., Li E., Spence M., Liapis S. C., Mallard W., Morse M., Swerdel M. R., D’Ecclessis M. F., Moore J. C., Lai V., Gong G., Yancopoulos G. D., Frendewey D., Kellis M., Hart R. P., Valenzuela D. M., Arlotta P., Rinn J. L., Multiple knockout mouse models reveal lincRNAs are required for life and brain development. eLife 2, e01749 (2013).
55
Delmas V., van der Hoorn F., Mellström B., Jégou B., Sassone-Corsi P., Induction of CREM activator proteins in spermatids: Down-stream targets and implications for haploid germ cell differentiation. Mol. Endocrinol. 7, 1502–1514 (1993).
56
Godmann M., Katz J. P., Guillou F., Simoni M., Kaestner K. H., Behr R., Krüppel-like factor 4 is involved in functional differentiation of testicular Sertoli cells. Dev. Biol. 315, 552–566 (2008).
57
B. Hogan, R. Beddington, F. Costantini, E. Lacy, Manipulating the Mouse Embryo: A Laboratory Manual (Cold Spring Harbor Laboratory Press, New York, ed. 2, 1994).
58
McGraw S., Zhang J. X., Farag M., Chan D., Caron M., Konermann C., Oakes C. C., Mohan K. N., Plass C., Pastinen T., Bourque G., Chaillet J. R., Trasler J. M., Transient DNMT1 suppression reveals hidden heritable marks in the genome. Nucleic Acids Res. 43, 1485–1497 (2015).
59
Xi Y., Li W., BSMAP: Whole genome bisulfite sequence MAPping program. BMC Bioinformatics 10, 232 (2009).
60
Goodrich R., Johnson G., Krawetz S. A., The preparation of human spermatozoal RNA for clinical analysis. Arch. Androl. 53, 161–167 (2007).
61
Ritchie M. E., Phipson B., Wu D., Hu Y., Law C. W., Shi W., Smyth G. K., limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).
62
Huang W., Sherman B. T., Lempicki R. A., Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).
63
A. Alexa, J. Rahnenfuhrer, topGO: Enrichment analysis for Gene Ontology. R package version 2.10.0 (2010).
64
Heard E., Martienssen R. A., Transgenerational epigenetic inheritance: Myths and mechanisms. Cell 157, 95–109 (2014).
65
Carmell M. A., Girard A., van de Kant H. J., Bourc’his D., Bestor T. H., de Rooij D. G., Hannon G. J., MIWI2 is essential for spermatogenesis and repression of transposons in the mouse male germline. Dev. Cell 12, 503–514 (2007).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science
Volume 350 | Issue 6261
6 November 2015

Article versions

You are viewing the most recent version of this article.

Submission history

Received: 26 March 2015
Accepted: 18 September 2015
Published in print: 6 November 2015

Permissions

Request permissions for this article.

Acknowledgments

We thank M. Stadler, H. Royo, and D. Gaidatzis (FMI) for advice on computational data analysis; B. K. Hall (Dalhousie University) and J. Tanny and D. Bernard (McGill University) for advice; X. Giner for technical expertise; the Laboratoire de Transgénèse Center de Recherché, Centre Hospitalier de l’Université de Montréal, Canada, for microinjection and generation of the transgenic hKDM1A founders; Y. Shi (Harvard University) for providing full-length human KDM1 cDNA; T. Pastinen, G. Bourque, M. Caron, and platform personnel of the McGill Epigenomics Mapping and Data Coordinating Centers in the McGill University, as well as Génome Québec Innovation Centre, for setting up the RRBS sequencing data analysis pipeline; and F. Lefebvre for support with microarray analysis. This research was funded by the Canadian Institute of Health Research (S.K. and J.T.), Genome Quebec (S.K. and J.T.), the Reseau de Reproduction Quebecois, Fonds de Recherche du Québec – Nature et Technologies (FRQNT) (S.K. and J.T), Boehringer Ingelheim Fond (S.E.), Swiss National Science Foundation (grant 31003A_125386) (A.H.F.M.P.), and the Novartis Research Foundation (A.H.F.M.P.). The data reported in this paper are available at Gene Expression Omnibus: Microarray data, including both the CEL files and the transcript-level expression values, are under accession number GSE66052; ChIP-seq and nucleosome data are under accession number GSE55471. K.S., S.E., M.G., R.L., S.M., J.T., A.H.F.M.P., and S.K. conceived and designed the experiments. K.S., S.E., M.G., R.L., C.L., T.C., S.M., J.X., J.T., M.S., A.H.F.M.P., and S.K. performed the experiments and analyzed the data. M.H. provided advice on the data analysis and the manuscript. K.S., J.T., A.H.F.M.P., and S.K. wrote the manuscript. We declare no conflicts of interest that would prejudice the impartiality of this work.

Authors

Affiliations

Keith Siklenka*
Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
Serap Erkek*
Friedrich Miescher Institute for Biomedical Research (FMI), CH-4058 Basel, Switzerland.
Faculty of Sciences, University of Basel, Basel, Switzerland.
Present address: European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany.
Present address: German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
Maren Godmann
Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada.
Present address: Department of Cell Biology, Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, Jena, Germany.
Romain Lambrot
Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada.
Serge McGraw
Department of Pediatrics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
Christine Lafleur
Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada.
Tamara Cohen
Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada.
Jianguo Xia
Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada.
Institute of Parasitology, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada.
Matthew Suderman
MRC Integrative Epidemiology Unity, School of Social and Community Medicine, University of Bristol, Bristol, UK.
Michael Hallett
McGill Centre for Bioinformatics, School of Computer Science, Faculty of Science, McGill University, Montreal, Quebec, Canada.
Jacquetta Trasler
Department of Pediatrics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
Department of Human Genetics and Department of Pharmacology and Therapeutics, Research Institute of the McGill University Health Centre at the Montreal Children's Hospital, Montreal, Quebec, Canada.
Antoine H. F. M. Peters*, [email protected]
Friedrich Miescher Institute for Biomedical Research (FMI), CH-4058 Basel, Switzerland.
Faculty of Sciences, University of Basel, Basel, Switzerland.
Sarah Kimmins*, [email protected]
Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada.

Notes

*
These authors contributed equally to this work.
||
Present address: Department of Obstetrics and Gynecology, Faculty of Medicine, Sainte-Justine Hospital Research Centre, Université de Montréal, Montreal, Quebec, Canada.
Corresponding author. E-mail: [email protected] (S.K.); [email protected] (A.H.F.M.P.)

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. Transcriptome profiling of histone writers/erasers enzymes across spermatogenesis, mature sperm and pre-cleavage embryo: Implications in paternal epigenome transitions and inheritance mechanisms, Frontiers in Cell and Developmental Biology, 11, (2023).https://doi.org/10.3389/fcell.2023.1086573
    Crossref
  2. Epigenetic resetting in the human germ line entails histone modification remodeling, Science Advances, 9, 3, (2023)./doi/10.1126/sciadv.ade1257
    Abstract
  3. Sperm DNA damage: The possible link between obesity and male infertility, an update of the current literature, Andrology, (2023).https://doi.org/10.1111/andr.13409
    Crossref
  4. Inheritance of paternal lifestyles and exposures through sperm DNA methylation, Nature Reviews Urology, (2023).https://doi.org/10.1038/s41585-022-00708-9
    Crossref
  5. Inheritance of epigenetic DNA marks studied in new mouse model, Nature, 615, 7954, (800-802), (2023).https://doi.org/10.1038/d41586-023-00708-8
    Crossref
  6. Nucleosomes in mammalian sperm: conveying paternal epigenetic inheritance or subject to reprogramming between generations?, Current Opinion in Genetics & Development, 79, (102034), (2023).https://doi.org/10.1016/j.gde.2023.102034
    Crossref
  7. Mechanisms of transgenerational epigenetic inheritance: lessons from animal model organisms, Current Opinion in Genetics & Development, 79, (102024), (2023).https://doi.org/10.1016/j.gde.2023.102024
    Crossref
  8. Present and future challenges for the investigation of transgenerational epigenetic inheritance, Environment International, 172, (107776), (2023).https://doi.org/10.1016/j.envint.2023.107776
    Crossref
  9. Transgenerational inheritance of wing development defects in Drosophila melanogaster induced by cadmium, Ecotoxicology and Environmental Safety, 250, (114486), (2023).https://doi.org/10.1016/j.ecoenv.2022.114486
    Crossref
  10. A program of successive gene expression in mouse one-cell embryos, Cell Reports, 42, 2, (112023), (2023).https://doi.org/10.1016/j.celrep.2023.112023
    Crossref
  11. See more
Loading...

View Options

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

Purchase this issue in print

Buy a single issue of Science for just $15 USD.

View options

PDF format

Download this article as a PDF file

Download PDF

Full Text

FULL TEXT

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media