Sperm epigenetics and influence of environmental factors

Ida Donkin, Romain Barrès, Ida Donkin, Romain Barrès

Abstract

Background: Developmental programming of the embryo is controlled by genetic information but also dictated by epigenetic information contained in spermatozoa. Lifestyle and environmental factors not only influence health in one individual but can also affect the phenotype of the following generations. This is mediated via epigenetic inheritance i.e., gametic transmission of environmentally-driven epigenetic information to the offspring. Evidence is accumulating that preconceptional exposure to certain lifestyle and environmental factors, such as diet, physical activity, and smoking, affects the phenotype of the next generation through remodeling of the epigenetic blueprint of spermatozoa.

Scope of review: This review will summarize current knowledge about the different epigenetic signals in sperm that are responsive to environmental and lifestyle factors and are capable of affecting embryonic development and the phenotype of the offspring later in life.

Major conclusions: Like somatic cells, the epigenome of spermatozoa has proven to be dynamically reactive to a wide variety of environmental and lifestyle stressors. The functional consequence on embryogenesis and phenotype of the next generation remains largely unknown. However, strong evidence of environmentally-driven sperm-borne epigenetic factors, which are capable of altering the phenotype of the next generation, is emerging on a large scale.

Keywords: DNA methylation; Epigenetic; Epigenetic inheritance; Histone; Small RNA; Sperm; Spermatozoa.

Copyright © 2018 The Authors. Published by Elsevier GmbH.. All rights reserved.

Figures

Figure 1
Figure 1
Lifestyle and environmental influences across generations. Exercise in the F0 generation may induce epigenetic reprogramming of the oocyte (1), and/or change whole body physiology (2) which, if still persistent when a pregnancy occurs, may have consequences on the extracellular milieu in utero (3). The developing embryo could be exposed to the exercise effects, thereby affecting not only the F1 (the embryo itself) but also the primordial germ cells developing in the embryo. Primordial germ cells represent, in part, the second-generation offspring, or F2. Exercise in the F0 may also alter behavior and metabolism in the F1 to influence aerobic capacity or inclination to exercise in the F1, which in turn induces programming of the spermatozoa through serial programming. Alternatively, exercise in the F0 may stably reprogram gametes throughout generations (F0, F1, … ), leading to true transgenerational epigenetic inheritance. Likely, the F2 generation is an integration of all epigenetic reprogramming that occurs throughout ancestors.
Figure 2
Figure 2
Overview of epigenetic marks susceptible to be remodeled with environmental insult. A simplified secondary structure of the sperm genome is represented, with the histone-bound DNA fraction accounting for less than 15% of the genome. DNA methylation remodeling is enhanced at CG rich, histone-bound fractions in sperm and is also found at repetitive elements. The positioning of histone relative to protamines may also be regulated by environmental factors. Histone modifications at specific loci are also changed after nutritional stress. Expression of small RNA (sRNA) such as tRNA fragments (tRF), microRNA (miRNA) and PIWI-interacting RNA (piRNA) is affected by lifestyle or environmental stress.

References

    1. Noble D. Evolution evolves: physiology returns to centre stage. The Journal of Physiology. 2014;592(11):2237–2244.
    1. Pembrey M.E. Sex-specific, male-line transgenerational responses in humans. European Journal of Human Genetics. 2006;14(2):159–166.
    1. Carone B.R. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell. 2010;143(7):1084–1096.
    1. Ng S.F. Paternal high-fat diet consumption induces common changes in the transcriptomes of retroperitoneal adipose and pancreatic islet tissues in female rat offspring. FASEB Journal. 2014;28(4):1830–1841.
    1. Radford E.J. In utero effects. In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science. 2014;345(6198):1255903.
    1. de Castro Barbosa T. High-fat diet reprograms the epigenome of rat spermatozoa and transgenerationally affects metabolism of the offspring. Molecular Metabolism. 2016;5(3):184–197.
    1. Denham J. Genome-wide sperm DNA methylation changes after 3 months of exercise training in humans. Epigenomics. 2015;7(5):717–731.
    1. Donkin I. Obesity and bariatric surgery drive epigenetic variation of spermatozoa in humans. Cell Metabolism. 2016;23(2):369–378.
    1. Ingerslev L.R. Endurance training remodels sperm-borne small RNA expression and methylation at neurological gene hotspots. Clinical Epigenetics. 2018
    1. Dias B.G. Parental olfactory experience influences behavior and neural structure in subsequent generations. Nature Neuroscience. 2014;17(1):89–96.
    1. Manikkam M. Plastics derived endocrine disruptors (BPA, DEHP and DBP) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations. PLoS One. 2013;8(1):e55387.
    1. McPherson N.O. Preconception diet or exercise intervention in obese fathers normalizes sperm microRNA profile and metabolic syndrome in female offspring. American Journal of Physiology-Endocrinology and Metabolism. 2015;308(9):E805–E821.
    1. Ge Z.J. Maternal diabetes causes alterations of DNA methylation statuses of some imprinted genes in murine oocytes. Biology of Reproduction. 2013;88(5):117.
    1. Ge Z.J. DNA methylation in oocytes and liver of female mice and their offspring: effects of high-fat-diet-induced obesity. Environmental Health Perspectives. 2014;122(2):159–164.
    1. Stilling R.M. Microbial genes, brain & behaviour - epigenetic regulation of the gut-brain axis. Genes, Brain and Behavior. 2014;13(1):69–86.
    1. Curley J.P. Epigenetics and the origins of paternal effects. Hormones and Behavior. 2011;59(3):306–314.
    1. Chen Q. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science. 2016;351(6271):397–400.
    1. Gapp K. Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nature Neuroscience. 2014;17(5):667–669.
    1. Grandjean V. RNA-mediated paternal heredity of diet-induced obesity and metabolic disorders. Scientific Reports. 2015;5:18193.
    1. Sharma U. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science. 2016;351(6271):391–396.
    1. Ventura-Junca P. In vitro fertilization (IVF) in mammals: epigenetic and developmental alterations. Scientific and bioethical implications for IVF in humans. Biological Research. 2015;48:68.
    1. Jaenisch R. DNA methylation and imprinting: why bother? Trends in Genetics. 1997;13(8):323–329.
    1. Bird A.P. Methylation-induced repression–belts, braces, and chromatin. Cell. 1999;99(5):451–454.
    1. Ito S. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010;466(7310):1129–1133.
    1. Tahiliani M. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science. 2009;324(5929):930–935.
    1. Woodcock D.M. The majority of methylated deoxycytidines in human DNA are not in the CpG dinucleotide. Biochemical and Biophysical Research Communications. 1987;145(2):888–894.
    1. Lister R. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. 2009;462(7271):315–322.
    1. Yan J. Evidence for non-CpG methylation in mammals. Experimental Cell Research. 2011;317(18):2555–2561.
    1. Barres R. Non-CpG methylation of the PGC-1alpha promoter through DNMT3B controls mitochondrial density. Cell Metabolism. 2009;10(3):189–198.
    1. Ichiyanagi T. Accumulation and loss of asymmetric non-CpG methylation during male germ-cell development. Nucleic Acids Research. 2013;41(2):738–745.
    1. Messerschmidt D.M. DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes and Development. 2014;28(8):812–828.
    1. Seisenberger S. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Molecular Cell. 2012;48(6):849–862.
    1. Tang W.W. Specification and epigenetic programming of the human germ line. Nature Reviews Genetics. 2016;17(10):585–600.
    1. Kobayashi H. High-resolution DNA methylome analysis of primordial germ cells identifies gender-specific reprogramming in mice. Genome Research. 2013;23(4):616–627.
    1. Gkountela S. DNA demethylation dynamics in the human prenatal germline. Cell. 2015;161(6):1425–1436.
    1. Wang X. Ultra-performance liquid chromatography/tandem mass spectrometry for accurate quantification of global DNA methylation in human sperms. Journal of Chromatography. B. 2011;879(19):1647–1652. Analytical Technologies in the Biomedical and Life Sciences.
    1. Molaro A. Sperm methylation profiles reveal features of epigenetic inheritance and evolution in primates. Cell. 2011;146(6):1029–1041.
    1. Ehrlich M. Amount and distribution of 5-methylcytosine in human DNA from different types of tissues of cells. Nucleic Acids Research. 1982;10(8):2709–2721.
    1. Simar D. DNA methylation is altered in B and NK lymphocytes in obese and type 2 diabetic human. Metabolism. 2014;63(9):1188–1197.
    1. Tang W.W. A unique gene regulatory network resets the human germline epigenome for development. Cell. 2015;161(6):1453–1467.
    1. Feinberg A.P. Evolution in health and medicine Sackler colloquium: stochastic epigenetic variation as a driving force of development, evolutionary adaptation, and disease. Proceedings of the National Academy of Sciences of the United States. 2010;107(Suppl 1):1757–1764.
    1. Feinberg J.I. Paternal sperm DNA methylation associated with early signs of autism risk in an autism-enriched cohort. International Journal of Epidemiology. 2015;44(4):1199–1210.
    1. Kerkel K. Genomic surveys by methylation-sensitive SNP analysis identify sequence-dependent allele-specific DNA methylation. Nature Genetics. 2008;40(7):904–908.
    1. Gertz J. Analysis of DNA methylation in a three-generation family reveals widespread genetic influence on epigenetic regulation. PLoS Genetics. 2011;7(8):e1002228.
    1. You J.S. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell. 2012;22(1):9–20.
    1. Wei Y. Paternally induced transgenerational inheritance of susceptibility to diabetes in mammals. Proceedings of the National Academy of Sciences of the United States. 2014;111(5):1873–1878.
    1. Shea J.M. Genetic and epigenetic variation, but not diet, shape the sperm methylome. Developmental Cell. 2015;35(6):750–758.
    1. Ost A. Paternal diet defines offspring chromatin state and intergenerational obesity. Cell. 2014;159(6):1352–1364.
    1. Ward W.S. DNA packaging and organization in mammalian spermatozoa: comparison with somatic cells. Biology of Reproduction. 1991;44(4):569–574.
    1. Barral S. Histone variant H2A.L.2 guides transition protein-dependent protamine assembly in male germ cells. Molecules and Cells. 2017;66(1):89–101. e108.
    1. Rousseaux S. Histone acylation beyond acetylation: terra incognita in chromatin biology. Cell Journal. 2015;17(1):1–6.
    1. Goudarzi A. Dynamic competing histone H4 K5K8 acetylation and butyrylation are hallmarks of highly active gene promoters. Molecules and Cells. 2016;62(2):169–180.
    1. Gaucher J. From meiosis to postmeiotic events: the secrets of histone disappearance. FEBS Journal. 2010;277(3):599–604.
    1. Arpanahi A. Endonuclease-sensitive regions of human spermatozoal chromatin are highly enriched in promoter and CTCF binding sequences. Genome Research. 2009;19(8):1338–1349.
    1. Erkek S. Molecular determinants of nucleosome retention at CpG-rich sequences in mouse spermatozoa. Nature Structural and Molecular Biology. 2013;20(7):868–875.
    1. Hammoud S.S. Genome-wide analysis identifies changes in histone retention and epigenetic modifications at developmental and imprinted gene loci in the sperm of infertile men. Human Reproduction. 2011;26(9):2558–2569.
    1. Carone B.R. High-resolution mapping of chromatin packaging in mouse embryonic stem cells and sperm. Developmental Cell. 2014;30(1):11–22.
    1. Samans B. Uniformity of nucleosome preservation pattern in Mammalian sperm and its connection to repetitive DNA elements. Developmental Cell. 2014;30(1):23–35.
    1. Kurimoto K. Mechanism and reconstitution in vitro of germ cell development in mammals. Cold Spring Harbor Symposia on Quantitative Biology. 2015;80:147–154.
    1. van der Heijden G.W. Sperm-derived histones contribute to zygotic chromatin in humans. BMC Developmental Biology. 2008;8:34.
    1. Teperek M. Sperm is epigenetically programmed to regulate gene transcription in embryos. Genome Research. 2016;26(8):1034–1046.
    1. Yamauchi Y. Paternal pronuclear DNA degradation is functionally linked to DNA replication in mouse oocytes. Biology of Reproduction. 2007;77(3):407–415.
    1. Ward W.S. Function of sperm chromatin structural elements in fertilization and development. Molecular Human Reproduction. 2010;16(1):30–36.
    1. Bjorndahl L. Sequence of ejaculation affects the spermatozoon as a Carrier and its message. Reproductive BioMedicine Online. 2003;7(4):440–448.
    1. Bjorndahl L. Human sperm chromatin stabilization: a proposed model including zinc bridges. Molecular Human Reproduction. 2010;16(1):23–29.
    1. Siklenka K. Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science. 2015;350(6261):aab2006.
    1. Zheng H. Resetting epigenetic memory by reprogramming of histone modifications in mammals. Molecules and Cells. 2016;63(6):1066–1079.
    1. Johnson G.D. Cleavage of rRNA ensures translational cessation in sperm at fertilization. Molecular Human Reproduction. 2011;17(12):721–726.
    1. Goodrich R.J. Isolating mRNA and small noncoding RNAs from human sperm. Methods in Molecular Biology. 2013;927:385–396.
    1. Ostermeier G.C. Reproductive biology: delivering spermatozoan RNA to the oocyte. Nature. 2004;429(6988):154.
    1. Sone Y. Nuclear translocation of phospholipase C-zeta, an egg-activating factor, during early embryonic development. Biochemical and Biophysical Research Communications. 2005;330(3):690–694.
    1. Rassoulzadegan M. RNA-mediated non-mendelian inheritance of an epigenetic change in the mouse. Nature. 2006;441(7092):469–474.
    1. Deng W. miwi, a murine homolog of piwi, encodes a cytoplasmic protein essential for spermatogenesis. Developmental Cell. 2002;2(6):819–830.
    1. Ashe A. piRNAs can trigger a multigenerational epigenetic memory in the germline of C. elegans. Cell. 2012;150(1):88–99.
    1. Brennecke J. An epigenetic role for maternally inherited piRNAs in transposon silencing. Science. 2008;322(5906):1387–1392.
    1. Lau J. CART in the regulation of appetite and energy homeostasis. Frontiers in Neuroscience. 2014;8:313.
    1. Castella S. Ilf3 and NF90 functions in RNA biology. Wiley Interdisciplinary Reviews: RNA. 2015;6(2):243–256.
    1. Jiang L.Q. Autocrine role of interleukin-13 on skeletal muscle glucose metabolism in type 2 diabetic patients involves microRNA let-7. American Journal of Physiology-Endocrinology and Metabolism. 2013;305(11):E1359–E1366.
    1. Zhu H. The Lin28/let-7 axis regulates glucose metabolism. Cell. 2011;147(1):81–94.
    1. Garcia-Lopez J. Global characterization and target identification of piRNAs and endo-siRNAs in mouse gametes and zygotes. Biochimica et Biophysica Acta. 2014;1839(6):463–475.
    1. Bartel D.P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–297.
    1. Liu W.M. Sperm-borne microRNA-34c is required for the first cleavage division in mouse. Proceedings of the National Academy of Sciences of the United States. 2012;109(2):490–494.
    1. Grandjean V. The miR-124-Sox9 paramutation: RNA-mediated epigenetic control of embryonic and adult growth. Development. 2009;136(21):3647–3655.
    1. Wagner K.D. RNA induction and inheritance of epigenetic cardiac hypertrophy in the mouse. Developmental Cell. 2008;14(6):962–969.
    1. Cropley J.E. Male-lineage transmission of an acquired metabolic phenotype induced by grand-paternal obesity. Molecular Metabolism. 2016;5(8):699–708.
    1. Rodgers A.B. Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress. Proceedings of the National Academy of Sciences of the United States. 2015;112(44):13699–13704.
    1. Alm P.S. Grandpaternal-induced transgenerational dietary reprogramming of the unfolded protein response in skeletal muscle. Molecular Metabolism. 2017;6(7):621–630.
    1. Krawetz S.A. A survey of small RNAs in human sperm. Human Reproduction. 2011;26(12):3401–3412.
    1. Nagano T. The Air noncoding RNA epigenetically silences transcription by targeting G9a to chromatin. Science. 2008;322(5908):1717–1720.
    1. Pandey R.R. Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Molecular Cell. 2008;32(2):232–246.
    1. Benetti R. A mammalian microRNA cluster controls DNA methylation and telomere recombination via Rbl2-dependent regulation of DNA methyltransferases. Nature Structural and Molecular Biology. 2008;15(9):998.
    1. Lai F. Where long noncoding RNAs meet DNA methylation. Cell Research. 2014;24(3):263–264.
    1. Li L.C. Chromatin remodeling by the small RNA machinery in mammalian cells. Epigenetics. 2014;9(1):45–52.
    1. Bird A. A fraction of the mouse genome that is derived from islands of nonmethylated, CpG-rich DNA. Cell. 1985;40(1):91–99.
    1. Cohen N.M. Primate CpG islands are maintained by heterogeneous evolutionary regimes involving minimal selection. Cell. 2011;145(5):773–786.
    1. Shen J.C. The rate of hydrolytic deamination of 5-methylcytosine in double-stranded DNA. Nucleic Acids Research. 1994;22(6):972–976.
    1. Waddington C.H. The epigenotype. 1942. International Journal of Epidemiology. 2012;41(1):10–13.
    1. Leisegang K. Obesity is associated with increased seminal insulin and leptin alongside reduced fertility parameters in a controlled male cohort. Reproductive Biology and Endocrinology. 2014;12:34.
    1. Fariello R.M. Effect of smoking on the functional aspects of sperm and seminal plasma protein profiles in patients with varicocele. Human Reproduction. 2012;27(11):3140–3149.
    1. Marques C.J. DNA methylation imprinting marks and DNA methyltransferase expression in human spermatogenic cell stages. Epigenetics. 2011;6(11):1354–1361.
    1. Ni K. TET enzymes are successively expressed during human spermatogenesis and their expression level is pivotal for male fertility. Human Reproduction. 2016;31(7):1411–1424.
    1. Pattamaprapanont P. Muscle contraction induces acute hydroxymethylation of the exercise-responsive gene Nr4a3. Frontiers in Endocrinology. 2016;7:165.
    1. Vojta A. Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Research. 2016;44(12):5615–5628.
    1. Liu X.S. Editing DNA methylation in the mammalian genome. Cell. 2016;167(1):233–247. e217.

Source: PubMed

3
Abonnere