Environmental Impact on Male (In)Fertility via Epigenetic Route
Abstract
:1. Introduction: An Overview of Epigenetic Mechanisms
2. Epigenetic Signature of Male Germline
2.1. Epigenetic Mechanisms during Germ Cell Progression
2.1.1. DNA Methylation
2.1.2. Chromatin Remodeling
2.1.3. NcRNAs
2.2. Epigenetic Mechanisms in Mature SPZ
2.3. Alterations of Epigenetic Mechanisms Cause the Spermatogenic Failure
3. Epigenetic Mechanisms Are Sensitive to Environment and Lifestyle
3.1. Environment Induced Epigenetic Modifications Targeting Spermatogenesis
3.2. Sperm Epigenome is Sensitive to Environmental Exposure and Conveys Environmentally Induced Epimutations Across Generations
3.3. Diet-Induced Epigenetic Modifications Targeting Germ Cells. SPZ as Carriers of Diet-Induced Epimutations Across Generations
4. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Mima, M.; Greenwald, D.; Ohlander, S. Environmental Toxins and Male Fertility. Curr. Urol. Rep. 2018, 19, 50. [Google Scholar] [CrossRef] [PubMed]
- Day, J.; Savani, S.; Krempley, B.D.; Nguyen, M.; Kitlinska, J.B. Influence of paternal preconception exposures on their offspring: Through epigenetics to phenotype. Am. J. Stem Cells 2016, 5, 11–18. [Google Scholar] [PubMed]
- Hotchkiss, R.D. The quantitative separation of purines, pyrimidines, and nucleosides by paper chromatography. J. Biol. Chem. 1948, 175, 315–332. [Google Scholar] [PubMed]
- Penn, N.W.; Suwalski, R.; O’Riley, C.; Bojanowski, K.; Yura, R. The presence of 5-hydroxymethylcytosine in animal deoxyribonucleic acid. Biochem. J. 1972, 126, 781–790. [Google Scholar] [CrossRef] [Green Version]
- Compere, S.J.; Palmiter, R.D. DNA methylation controls the inducibility of the mouse metallothionein-I gene in lymphoid cells. Cell 1981, 25, 233–240. [Google Scholar] [CrossRef]
- Patil, V.; Ward, R.L.; Hesson, L.B. The evidence for functional non-CpG methylation in mammalian cells. Epigenetics 2014, 9, 823–828. [Google Scholar] [CrossRef] [Green Version]
- Bestor, T.H.; Edwards, J.R.; Boulard, M. Notes on the role of dynamic DNA methylation in mammalian development. Proc. Natl. Acad. Sci. USA 2015, 112, 6796–6799. [Google Scholar] [CrossRef] [Green Version]
- Kelly, T.K.; Liu, Y.; Lay, F.D.; Liang, G.; Berman, B.P.; Jones, P.A. Genome-wide mapping of nucleosome positioning and DNA methylation within individual DNA molecules. Genome Res. 2012, 22, 2497–2506. [Google Scholar] [CrossRef] [Green Version]
- Tahiliani, M.; Koh, K.P.; Shen, Y.; Pastor, W.A.; Bandukwala, H.; Brudno, Y.; Agarwal, S.; Iyer, L.M.; Liu, D.R.; Aravind, L.; et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009, 324, 930–935. [Google Scholar] [CrossRef] [Green Version]
- Morales, V.; Giamarchi, C.; Chailleux, C.; Moro, F.; Marsaud, V.; Le Ricousse, S.; Richard-Foy, H. Chromatin structure and dynamics: Functional implications. Biochimie 2001, 83, 1029–1039. [Google Scholar] [CrossRef]
- Allfrey, V.G.; Faulkner, R.; Mirsky, A.E. Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc. Natl. Acad. Sci. USA 1964, 51, 786–794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lawrence, M.; Daujat, S.; Schneider, R. lateral thinking: How histone modifications regulate gene expression. Trends Genet. 2016, 32, 42–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santos-Rosa, H.; Schneider, R.; Bannister, A.J.; Sherriff, J.; Bernstein, B.E.; Emre, N.C.T.; Schreiber, S.L.; Mellor, J.; Kouzarides, T. Active genes are tri-methylated at K4 of histone H3. Nature 2002, 419, 407–411. [Google Scholar] [CrossRef] [PubMed]
- Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef] [PubMed]
- Khawar, M.B.; Mehmood, R.; Roohi, N. MicroRNAs: Recent insights towards their role in male infertility and reproductive cancers. Bosn. J. Basic Med. Sci. 2019, 19, 31–42. [Google Scholar] [CrossRef] [PubMed]
- Panni, S.; Lovering, R.C.; Porras, P.; Orchard, S. Non-coding RNA regulatory networks. Biochim. Biophys. Acta-Gene Regul. Mech. 2020, 1863, 194417. [Google Scholar] [CrossRef]
- Hurley, D.; Araki, H.; Tamada, Y.; Dunmore, B.; Sanders, D.; Humphreys, S.; Affara, M.; Imoto, S.; Yasuda, K.; Tomiyasu, Y.; et al. Gene network inference and visualization tools for biologists: Application to new human transcriptome datasets. Nucleic Acids Res. 2012, 40, 2377–2398. [Google Scholar] [CrossRef]
- Lillycrop, K.A.; Hoile, S.P.; Grenfell, L.; Burdge, G.C. DNA methylation, ageing and the influence of early life nutrition. Proc. Nutr. Soc. 2014, 73, 413–421. [Google Scholar] [CrossRef] [Green Version]
- Choi, S.W.; Friso, S. Epigenetics: A new bridge between nutrition and health. Adv. Nutr. 2010, 1, 8–16. [Google Scholar] [CrossRef]
- Huang, B.; Jiang, C.; Zhang, R. Epigenetics: The language of the cell? Epigenomics 2014, 6, 73–88. [Google Scholar] [CrossRef] [Green Version]
- Xavier, M.J.; Roman, S.D.; Aitken, R.J.; Nixon, B. Transgenerational inheritance: How impacts to the epigenetic and genetic information of parents affect offspring health. Hum. Reprod. Update 2019, 25, 519–541. [Google Scholar] [CrossRef] [PubMed]
- Champroux, A.; Cocquet, J.; Henry-Berger, J.; Drevet, J.R.; Kocer, A. A decade of exploring the mammalian sperm epigenome: Paternal epigenetic and transgenerational inheritance. Front. Cell Dev. Biol. 2018, 6, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rando, O.J. Intergenerational transfer of epigenetic information in sperm. Cold Spring Harb. Perspect. Med. 2016, 6, a022988. [Google Scholar] [CrossRef] [Green Version]
- Sharma, U. Paternal contributions to offspring health: Role of sperm small RNAs in intergenerational transmission of epigenetic information. Front. Cell Dev. Biol. 2019, 7, 215. [Google Scholar] [CrossRef] [PubMed]
- Prokopuk, L.; Western, P.S.; Stringer, J.M. Transgenerational epigenetic inheritance: Adaptation through the germline epigenome? Epigenomics 2015, 7, 829–846. [Google Scholar] [CrossRef] [Green Version]
- Messerschmidt, D.M.; Knowles, B.B.; Solter, D. DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev. 2014, 28, 812–828. [Google Scholar] [CrossRef] [Green Version]
- De Kretser, D.M.; Loveland, K.; O’Bryan, M. Spermatogenesis. In Endocrinology: Adult and Pediatric; Elsevier: Philadelphia, PA, USA, 2016; pp. 2325–2353. ISBN 9780323189071. [Google Scholar]
- Gold, H.B.; Jung, Y.H.; Corces, V.G. Not just heads and tails: The complexity of the sperm epigenome. J. Biol. Chem. 2018, 293, 13815–13820. [Google Scholar] [CrossRef] [Green Version]
- Oakes, C.C.; La Salle, S.; Smiraglia, D.J.; Robaire, B.; Trasler, J.M. Developmental acquisition of genome-wide DNA methylation occurs prior to meiosis in male germ cells. Dev. Biol. 2007, 307, 368–379. [Google Scholar] [CrossRef] [Green Version]
- Marques, C.J.; Pinho, M.J.; Carvalho, F.; Bièche, I.; Barros, A.; Sousa, M. DNA methylation imprinting marks and DNA methyltransferase expression in human spermatogenic cell stages. Epigenetics 2011, 6, 1354–1361. [Google Scholar] [CrossRef] [Green Version]
- Davis, T.L. The H19 methylation imprint is erased and re-established differentially on the parental alleles during male germ cell development. Hum. Mol. Genet. 2000, 9, 2885–2894. [Google Scholar] [CrossRef]
- Lucifero, D.; Mertineit, C.; Clarke, H.J.; Bestor, T.H.; Trasler, J.M. Methylation Dynamics of Imprinted Genes in Mouse Germ Cells. Genomics 2002, 79, 530–538. [Google Scholar] [CrossRef] [PubMed]
- Arnaud, P. Genomic imprinting in germ cells: Imprints are under control. Reproduction 2010, 140, 411–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walsh, C.P.; Chaillet, J.R.; Bestor, T.H. Transcription of IAP endogenous retroviruses is constrained by cytosine methylation. Nat. Genet. 1998, 20, 116–117. [Google Scholar] [CrossRef] [PubMed]
- Gromoll, J.; Kläver, R. Bringing epigenetics into the diagnostics of the andrology laboratory: Challenges and perspectives. Asian J. Androl. 2014, 16, 669. [Google Scholar] [CrossRef]
- Carrell, D.T.; Emery, B.R.; Hammoud, S. Altered protamine expression and diminished spermatogenesis: What is the link? Hum. Reprod. Update 2007, 13, 313–327. [Google Scholar] [CrossRef] [Green Version]
- Boissonnas, C.C.; Jouannet, P.; Jammes, H. Epigenetic disorders and male subfertility. Fertil. Steril. 2013, 99, 624–631. [Google Scholar] [CrossRef]
- Sonnack, V.; Failing, K.; Bergmann, M.; Steger, K. Expression of hyperacetylated histone H4 during normal and impaired human spermatogenesis. Andrologia 2002, 34, 384–390. [Google Scholar] [CrossRef]
- Balhorn, R.; Cosman, M.; Thornton, K.; Krishnan, V.V.; Corzett, M.; Bench, G.; Kramer, C.; Lee, J.I.; Hud, N.V.; Allen, M.; et al. Protamine mediated condensation of DNA in mammalian sperm. In Male Gamete: From Basic Science to Clinical Applications; Cache River Press: St. Louis, MO, USA, 1999. [Google Scholar]
- Carrell, D.T.; Liu, L. Altered protamine 2 expression is uncommon in donors of known fertility, but common among men with poor fertilizing capacity, and may reflect other abnormalities, of spermiogenesis. J. Androl. 2001, 22, 604–610. [Google Scholar] [CrossRef]
- Oliva, R. Protamines and male infertility. Hum. Reprod. Update 2006, 12, 417–435. [Google Scholar] [CrossRef] [Green Version]
- Chevaillier, P.; Mauro, N.; Feneux, D.; Jouannet, P.; David, G. Anomalous Protein Complement of Sperm Nuclei in Some Infertile Men. Lancet 1987, 330, 806–807. [Google Scholar] [CrossRef]
- Balhorn, R.; Reed, S.; Tanphaichitr, N. Aberrant protamine 1/protamine 2 ratios in sperm of infertile human males. Experientia 1988, 44, 52–55. [Google Scholar] [CrossRef] [PubMed]
- Belokopytova, I.A.; Kostyleva, E.I.; Tomilin, A.N.; Vorob’ev, V.I. Human male infertility may be due to a decrease of the protamine P2 content in sperm chromatin. Mol. Reprod. Dev. 1993, 34, 53–57. [Google Scholar] [CrossRef] [PubMed]
- De Yebra, L.; Ballescá, J.L.; Vanrell, J.A.; Corzett, M.; Balhorn, R.; Oliva, R. Detection of P2 precursors in the sperm cells of infertile patients who have reduced protamine P2 levels. Fertil. Steril. 1998, 69, 755–759. [Google Scholar] [CrossRef]
- Aoki, V.W.; Liu, L.; Carrell, D.T. Identification and evaluation of a novel sperm protamine abnormality in a population of infertile males. Hum. Reprod. 2005, 20, 1298–1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. 2001, 28, 82–86. [Google Scholar] [CrossRef]
- Papaioannou, M.D.; Nef, S. MicroRNAs in the testis: Building up male fertility. J. Androl. 2010, 31, 26–33. [Google Scholar] [CrossRef]
- Shomron, N.; Levy, C. MicroRNA-Biogenesis and Pre-mRNA Splicing Crosstalk. J. Biomed. Biotechnol. 2009, 2009, 1–6. [Google Scholar] [CrossRef]
- De Mateo, S.; Sassone-Corsi, P. Regulation of spermatogenesis by small non-coding RNAs: Role of the germ granule. Semin. Cell Dev. Biol. 2014, 29, 84–92. [Google Scholar] [CrossRef] [Green Version]
- Watanabe, T.; Chuma, S.; Yamamoto, Y.; Kuramochi-Miyagawa, S.; Totoki, Y.; Toyoda, A.; Hoki, Y.; Fujiyama, A.; Shibata, T.; Sado, T.; et al. MITOPLD is a mitochondrial protein essential for nuage formation and piRNA biogenesis in the mouse germline. Dev. Cell 2011, 20, 364–375. [Google Scholar] [CrossRef] [Green Version]
- Lian, J.; Zhang, X.; Tian, H.; Liang, N.; Wang, Y.; Liang, C.; Li, X.; Sun, F. Altered microRNA expression in patients with non-obstructive azoospermia. Reprod. Biol. Endocrinol. 2009, 7, 13. [Google Scholar] [CrossRef] [Green Version]
- Abu-Halima, M.; Hammadeh, M.; Schmitt, J.; Leidinger, P.; Keller, A.; Meese, E.; Backes, C. Altered microRNA expression profiles of human spermatozoa in patients with different spermatogenic impairments. Fertil. Steril. 2013, 99, 1249–1255.e16. [Google Scholar] [CrossRef] [PubMed]
- Abu-Halima, M.; Backes, C.; Leidinger, P.; Keller, A.; Lubbad, A.M.; Hammadeh, M.; Meese, E. MicroRNA expression profiles in human testicular tissues of infertile men with different histopathologic patterns. Fertil. Steril. 2014, 101, 78–86.e2. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Liu, Y.; Su, D.; Yang, Y.; Bai, G.; Tao, D.; Ma, Y.; Zhang, S. A single nucleotide polymorphism in a miR-1302 binding site in CGA increases the risk of idiopathic male infertility. Fertil. Steril. 2011, 96, 34–39.e7. [Google Scholar] [CrossRef] [PubMed]
- Qin, Y.; Xia, Y.; Wu, W.; Han, X.; Lu, C.; Ji, G.; Chen, D.; Wang, H.; Song, L.; Wang, S.; et al. Genetic variants in microRNA biogenesis pathway genes are associated with semen quality in a Han-Chinese population. Reprod. Biomed. Online 2012, 24, 454–461. [Google Scholar] [CrossRef] [Green Version]
- Gu, A.; Ji, G.; Shi, X.; Long, Y.; Xia, Y.; Song, L.; Wang, S.; Wang, X. Genetic variants in Piwi-interacting RNA pathway genes confer susceptibility to spermatogenic failure in a Chinese population. Hum. Reprod. 2010, 25, 2955–2961. [Google Scholar] [CrossRef] [Green Version]
- Friemel, C.; Ammerpohl, O.; Gutwein, J.; Schmutzler, A.G.; Caliebe, A.; Kautza, M.; von Otte, S.; Siebert, R.; Bens, S. Array-based DNA methylation profiling in male infertility reveals allele-specific DNA methylation in PIWIL1 and PIWIL2. Fertil. Steril. 2014, 101, 1097–1103.e1. [Google Scholar] [CrossRef]
- Lin, X.; Han, M.; Cheng, L.; Chen, J.; Zhang, Z.; Shen, T.; Wang, M.; Wen, B.; Ni, T.; Han, C. Expression dynamics, relationships, and transcriptional regulations of diverse transcripts in mouse spermatogenic cells. RNA Biol. 2016, 13, 1011–1024. [Google Scholar] [CrossRef] [Green Version]
- Zhou, T.; Xie, X.; Li, M.; Shi, J.; Zhou, J.J.; Knox, K.S.; Wang, T.; Chen, Q.; Gu, W. Rat BodyMap transcriptomes reveal unique circular RNA features across tissue types and developmental stages. RNA 2018, 24, 1443–1456. [Google Scholar] [CrossRef] [Green Version]
- Dong, W.W.; Li, H.M.; Qing, X.R.; Huang, D.H.; Li, H.G. Identification and characterization of human testis derived circular RNAs and their existence in seminal plasma. Sci. Rep. 2016, 6, 39080. [Google Scholar] [CrossRef] [Green Version]
- Sun, J.; Lin, Y.; Wu, J. Long Non-Coding RNA Expression Profiling of Mouse Testis during Postnatal Development. PLoS ONE 2013, 8, e75750. [Google Scholar] [CrossRef]
- Bie, B.; Wang, Y.; Li, L.; Fang, H.; Liu, L.; Sun, J. Noncoding RNAs: Potential players in the self-renewal of mammalian spermatogonial stem cells. Mol. Reprod. Dev. 2018, 85, 720–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, L.; Wang, M.; Wang, M.; Wu, X.; Geng, L.; Xue, Y.; Wei, X.; Jia, Y.; Wu, X. A long non-coding RNA interacts with gfra1 and maintains survival of mouse spermatogonial stem cells. Cell Death Dis. 2016, 7. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.; Seo, H.H.; Lee, C.Y.; Lee, J.; Shin, S.; Kim, S.W.; Lim, S.; Hwang, K.C. Human Long Noncoding RNA Regulation of Stem Cell Potency and Differentiation. Stem Cells Int. 2017, 2017, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, C.Y.; Mruk, D.D. Regulation of spermiogenesis, spermiation and blood-testis barrier dynamics: Novel insights from studies on Eps8 and Arp3. Biochem. J. 2011, 435, 553–562. [Google Scholar] [CrossRef]
- Chianese, R.; Pierantoni, R. Sperm Cells, an Efficient Shuttle for the Intergenerational Epigenetic Memory. J. Pituit. Res. Treat. 2016, 1, 1–2. [Google Scholar]
- Horsthemke, B. A critical view on transgenerational epigenetic inheritance in humans. Nat. Commun. 2018, 9, 2973. [Google Scholar] [CrossRef] [Green Version]
- Rathke, C.; Barckmann, B.; Burkhard, S.; Jayaramaiah-Raja, S.; Roote, J.; Renkawitz-Pohl, R. Distinct functions of Mst77F and protamines in nuclear shaping and chromatin condensation during Drosophila spermiogenesis. Eur. J. Cell Biol. 2010, 89, 326–338. [Google Scholar] [CrossRef]
- Teperek, M.; Miyamoto, K. Nuclear reprogramming of sperm and somatic nuclei in eggs and oocytes. Reprod. Med. Biol. 2013, 12, 133–149. [Google Scholar] [CrossRef] [Green Version]
- Malo, A.F.; Gomendio, M.; Garde, J.; Lang-Lenton, B.; Soler, A.J.; Roldan, E.R.S. Sperm design and sperm function. Biol. Lett. 2006, 2, 246–249. [Google Scholar] [CrossRef] [Green Version]
- Govin, J.; Caron, C.; Lestrat, C.; Rousseaux, S.; Khochbin, S. The role of histones in chromatin remodelling during mammalian spermiogenesis. Eur. J. Biochem. 2004, 271, 3459–3469. [Google Scholar] [CrossRef]
- Khalil, A.M.; Wahlestedt, C. Epigenetic mechanisms of gene regulation during mammalian spermatogenesis. Epigenetics 2008, 3, 21–27. [Google Scholar] [CrossRef]
- Chioccarelli, T.; Pierantoni, R.; Manfrevola, F.; Porreca, V.; Fasano, S.; Chianese, R.; Cobellis, G. Histone post-translational modifications and CircRNAs in mouse and human spermatozoa: Potential epigenetic marks to assess human sperm Quality. J. Clin. Med. 2020, 9, 640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Erkek, S.; Hisano, M.; Liang, C.Y.; Gill, M.; Murr, R.; Dieker, J.; Schübeler, D.; Vlag, J.; Van Der Stadler, M.B.; Peters, A.H.F.M. Molecular determinants of nucleosome retention at CpG-rich sequences in mouse spermatozoa. Nat. Struct. Mol. Biol. 2013, 20, 868–875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fernandez-Capetillo, O.; Mahadevaiah, S.K.; Celeste, A.; Romanienko, P.J.; Camerini-Otero, R.D.; Bonner, W.M.; Manova, K.; Burgoyne, P.; Nussenzweig, A. H2AX is required for chromatin remodeling and inactivation of sex chromosomes in male mouse meiosis. Dev. Cell 2003, 4, 497–508. [Google Scholar] [CrossRef] [Green Version]
- Churikov, D.; Siino, J.; Svetlova, M.; Zhang, K.; Gineitis, A.; Morton Bradbury, E.; Zalensky, A. Novel human testis-specific histone H2B encoded by the interrupted gene on the X chromosome. Genomics 2004, 84, 745–756. [Google Scholar] [CrossRef]
- Palmer, D.K.; O’Day, K.; Margolis, R.L. The centromere specific histone CENP-A is selectively retained in discrete foci in mammalian sperm nuclei. Chromosoma 1990, 100, 32–36. [Google Scholar] [CrossRef]
- Usachenko, S.I.; Morton Bradbury, E. Histone-DNA contacts in structure/function relationships of nucleosomes as revealed by crosslinking. In Genetica; Springer: Dordrecht, The Netherlands, 1999; Volume 106, pp. 103–115. [Google Scholar]
- Rathke, C.; Baarends, W.M.; Awe, S.; Renkawitz-Pohl, R. Chromatin dynamics during spermiogenesis. Biochim. Biophys. Acta-Gene Regul. Mech. 2014, 1839, 155–168. [Google Scholar] [CrossRef] [Green Version]
- Rice, J.C.; Allis, C.D. Histone methylation versus histone acetylation: New insights into epigenetic regulation. Curr. Opin. Cell Biol. 2001, 13, 263–273. [Google Scholar] [CrossRef]
- Aoki, V.; Liu, L.; Jones, K.; Hatasaka, H.; Gibson, M.; Peterson, C.; Carrell, D. Sperm protamine 1/protamine 2 ratios are related to in vitro fertilization pregnancy rates and predictive of fertilization ability. Fertil. Steril. 2006, 86, 1408–1415. [Google Scholar] [CrossRef]
- Krejčí, J.; Stixová, L.; Pagáčová, E.; Legartová, S.; Kozubek, S.; Lochmanová, G.; Zdráhal, Z.; Sehnalová, P.; Dabravolski, S.; Hejátko, J.; et al. Post-translational modifications of histones in human sperm. J. Cell. Biochem. 2015, 116, 2195–2209. [Google Scholar] [CrossRef]
- 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 2009, 460, 473–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.; San Gabriel, M.; Zini, A. Sperm nuclear histone to protamine ratio in fertile and infertile men: Evidence of heterogeneous subpopulations of spermatozoa in the ejaculate. J. Androl. 2006, 27, 414–420. [Google Scholar] [CrossRef] [PubMed]
- Linnemann, A.K.; Krawetz, S.A. Maintenance of a functional higher order chromatin structure: The role of the nuclear matrix in normal and disease states. Gene Ther. Mol. Biol. 2009, 13, 231–243. [Google Scholar] [PubMed]
- Ward, W.S.; Coffey, D.S. DNA Packaging and organization in mammalian spermatozoa: Comparison with somatic cell. Biol. Reprod. 1991, 44, 569–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Popp, C.; Dean, W.; Feng, S.; Cokus, S.J.; Andrews, S.; Pellegrini, M.; Jacobsen, S.E.; Reik, W. Genome-wide erasure of DNA methylation in mouse primordial germ cells is affected by AID deficiency. Nature 2010, 463, 1101–1105. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, H.; Sakurai, T.; Imai, M.; Takahashi, N.; Fukuda, A.; Yayoi, O.; Sato, S.; Nakabayashi, K.; Hata, K.; Sotomaru, Y.; et al. Contribution of intragenic DNA methylation in mouse gametic DNA methylomes to establish Oocyte-specific heritable marks. PLoS Genet. 2012, 8, e1002440. [Google Scholar] [CrossRef] [Green Version]
- Guo, H.; Zhu, P.; Yan, L.; Li, R.; Hu, B.; Lian, Y.; Yan, J.; Ren, X.; Lin, S.; Li, J.; et al. The DNA methylation landscape of human early embryos. Nature 2014, 511, 606–610. [Google Scholar] [CrossRef]
- Molaro, A.; Hodges, E.; Fang, F.; Song, Q.; McCombie, W.R.; Hannon, G.J.; Smith, A.D. Sperm methylation profiles reveal features of epigenetic inheritance and evolution in primates. Cell 2011, 146, 1029–1041. [Google Scholar] [CrossRef] [Green Version]
- McLay, D.W.; Clarke, H.J. Remodelling the paternal chromatin at fertilization in mammals. Reproduction 2003, 125, 625–633. [Google Scholar] [CrossRef]
- Cui, X.; Jing, X.; Wu, X.; Yan, M.; Li, Q.; Shen, Y.; Wang, Z. DNA methylation in spermatogenesis and male infertility. Exp. Ther. Med. 2016, 12, 1973–1979. [Google Scholar] [CrossRef] [Green Version]
- Efimova, O.A.; Pendina, A.A.; Tikhonov, A.V.; Parfenyev, S.E.; Mekina, I.D.; Komarova, E.M.; Mazilina, M.A.; Daev, E.V.; Chiryaeva, O.G.; Galembo, I.A.; et al. Genome-wide 5-hydroxymethylcytosine patterns in human spermatogenesis are associated with semen quality. Oncotarget 2017, 8, 88294–88307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Waal, E.; Yamazaki, Y.; Ingale, P.; Bartolomei, M.; Yanagimachi, R.; McCarrey, J.R. Primary epimutations introduced during intracytoplasmic sperm injection (ICSI) are corrected by germline-specific epigenetic reprogramming. Proc. Natl. Acad. Sci. USA 2012, 109, 4163–4168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ge, S.Q.; Lin, S.L.; Zhao, Z.H.; Sun, Q.Y. Epigenetic dynamics and interplay during spermatogenesis and embryogenesis: Implications for male fertility and offspring health. Oncotarget 2017, 8, 53804–53818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benchaib, M.; Braun, V.; Ressnikof, D.; Lornage, J.; Durand, P.; Niveleau, A.; Guérin, J.F. Influence of global sperm DNA methylation on IVF results. Hum. Reprod. 2005, 20, 768–773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, D.; Ostermeier, G.C.; Krawetz, S.A. The controversy, potential and roles of spermatozoal RNA. Trends Mol. Med. 2005, 11, 156–163. [Google Scholar] [CrossRef] [PubMed]
- Lambard, S.; Galeraud-Denis, I.; Saunders, P.T.; Carreau, S. Human immature germ cells and ejaculated spermatozoa contain aromatase and oestrogen receptors. J. Mol. Endocrinol. 2004, 32, 279–289. [Google Scholar] [CrossRef] [Green Version]
- Ostermeier, G.C.; Miller, D.; Huntriss, J.D.; Diamond, M.P.; Krawetz, S.A. Reproductive biology: Delivering spermatozoan RNA to the oocyte. Nature. 2004, 429, 154. [Google Scholar] [CrossRef]
- Carreau, S.; Lambard, S.; Said, L.; Saad, A.; Galeraud-Denis, I. RNA dynamics of fertile and infertile spermatozoa. Biochem. Soc. Trans. 2007, 35, 634–636. [Google Scholar] [CrossRef]
- Bansal, S.K.; Gupta, N.; Sankhwar, S.N.; Rajender, S. Differential genes expression between fertile and infertile spermatozoa revealed by transcriptome analysis. PLoS ONE. 2015, 10, e0127007. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.; Yan, M.; Cao, Z.; Li, X.; Zhang, Y.; Shi, J.; Feng, G.H.; Peng, H.; Zhang, X.; Zhang, Y.; et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science 2016, 351, 397–400. [Google Scholar] [CrossRef] [Green Version]
- Chu, C.; Yu, L.; Wu, B.; Ma, L.; Gou, L.T.; He, M.; Guo, Y.; Li, Z.T.; Gao, W.; Shi, H.; et al. A sequence of 28S rRNA-derived small RNAs is enriched in mature sperm and various somatic tissues and possibly associates with inflammation. J. Mol. Cell Biol. 2017, 9, 256–259. [Google Scholar] [CrossRef] [PubMed]
- Chioccarelli, T.; Manfrevola, F.; Ferraro, B.; Sellitto, C.; Cobellis, G.; Migliaccio, M.; Fasano, S.; Pierantoni, R.; Chianese, R. Expression patterns of circular RNAs in high quality and poor quality human spermatozoa. Front. Endocrinol. 2019, 10. [Google Scholar] [CrossRef] [PubMed]
- Ragusa, M.; Barbagallo, D.; Chioccarelli, T.; Manfrevola, F.; Cobellis, G.; Di Pietro, C.; Brex, D.; Battaglia, R.; Fasano, S.; Ferraro, B.; et al. CircNAPEPLD is expressed in human and murine spermatozoa and physically interacts with oocyte miRNAs. RNA Biol. 2019, 16, 1237–1248. [Google Scholar] [CrossRef] [PubMed]
- Sullivan, R.; Saez, F. Epididymosomes, prostasomes, and liposomes: Their roles in mammalian male reproductive physiology. Reproduction 2013, 146, R21–R35. [Google Scholar] [CrossRef] [Green Version]
- Rybak-Wolf, A.; Stottmeister, C.; Glažar, P.; Jens, M.; Pino, N.; Hanan, M.; Behm, M.; Bartok, O.; Ashwal-Fluss, R.; Herzog, M.; et al. Circular RNAs in the mammalian brain are highly abundant, conserved, and dynamically expressed. Mol. Cell 2014, 58, 870–885. [Google Scholar] [CrossRef] [Green Version]
- Fan, X.; Zhang, X.; Wu, X.; Guo, H.; Hu, Y.; Tang, F.; Huang, Y. Single-cell RNA-seq transcriptome analysis of linear and circular RNAs in mouse preimplantation embryos. Genome Biol. 2015, 16, 148. [Google Scholar] [CrossRef] [Green Version]
- Dang, Y.; Yan, L.; Hu, B.; Fan, X.; Ren, Y.; Li, R.; Lian, Y.; Yan, J.; Li, Q.; Zhang, Y.; et al. Tracing the expression of circular RNAs in human pre-implantation embryos. Genome Biol. 2016, 17, 130. [Google Scholar] [CrossRef] [Green Version]
- Manfrevola, F.; Chioccarelli, T.; Cobellis, G.; Fasano, S.; Ferraro, B.; Sellitto, C.; Marella, G.; Pierantoni, R.; Chianese, R. CircRNA role and circRNA-dependent network (ceRNET) in asthenozoospermia. Front. Endocrinol. 2020, 11. [Google Scholar] [CrossRef]
- Okamoto, Y.; Morishita, J.; Tsuboi, K.; Tonai, T.; Ueda, N. Molecular Characterization of a Phospholipase D Generating Anandamide and Its Congeners. J. Biol. Chem. 2004, 279, 5298–5305. [Google Scholar] [CrossRef] [Green Version]
- Cobellis, G.; Cacciola, G.; Scarpa, D.; Meccariello, R.; Chianese, R.; Franzoni, M.F.; Mackie, K.; Pierantoni, R.; Fasano, S. Endocannabinoid system in frog and rodent testis: Type-1 cannabinoid receptor and fatty acid amide hydrolase activity in male germ cells. Biol. Reprod. 2006, 75, 82–89. [Google Scholar] [CrossRef] [Green Version]
- Fasano, S.; Meccariello, R.; Cobellis, G.; Chianese, R.; Cacciola, G.; Chioccarelli, T.; Pierantoni, R. The endocannabinoid system: An ancient signaling involved in the control of male fertility. Ann. N. Y. Acad. Sci. 2009, 1163, 112–124. [Google Scholar] [CrossRef]
- Chianese, R.; Ciaramella, V.; Scarpa, D.; Fasano, S.; Pierantoni, R.; Meccariello, R. Anandamide regulates the expression of GnRH1, GnRH2, and GnRH-Rs in frog testis. Am. J. Physiol.-Endocrinol. Metab. 2012, 303, E475–E487. [Google Scholar] [CrossRef]
- Grimaldi, P.; Di Giacomo, D.; Geremia, R. The endocannabinoid system and spermatogenesis. Front. Endocrinol. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
- Ciaramella, V.; Meccariello, R.; Chioccarelli, T.; Sirleto, M.; Fasano, S.; Pierantoni, R.; Chianese, R. Anandamide acts via kisspeptin in the regulation of testicular activity of the frog, Pelophylax esculentus. Mol. Cell. Endocrinol. 2016, 420, 75–84. [Google Scholar] [CrossRef]
- Castillo, J.; Amaral, A.; Oliva, R. Sperm nuclear proteome and its epigenetic potential. Andrology 2014, 2, 326–338. [Google Scholar] [CrossRef]
- Chatzimeletiou, K.; Morrison, E.E.; Prapas, N.; Prapas, Y.; Handyside, A.H. The centrosome and early embryogenesis: Clinical insights. Reprod. Biomed. Online 2008, 16, 485–491. [Google Scholar] [CrossRef]
- Baker, M.A.; Naumovski, N.; Hetherington, L.; Weinberg, A.; Velkov, T.; Aitken, R.J. Head and flagella subcompartmental proteomic analysis of human spermatozoa. Proteomics 2013, 13, 61–74. [Google Scholar] [CrossRef]
- Kierszenbaum, A.L.; Tres, L.L. RNA transcription and chromatin structure during meiotic and postmeiotic stages of spermatogenesis. Fed. Proc. 1978, 37, 2512–2516. [Google Scholar]
- Miller, D. Analysis and significance of messenger RNA in human ejaculated spermatozoa. Mol. Reprod. Dev. 2000, 56, 259–264. [Google Scholar] [CrossRef]
- Zhang, Y.; Shi, J.; Rassoulzadegan, M.; Tuorto, F.; Chen, Q. Sperm RNA code programmes the metabolic health of offspring. Nat. Rev. Endocrinol. 2019, 15, 489–498. [Google Scholar] [CrossRef] [Green Version]
- Bromfield, J.J. Seminal fluid and reproduction: Much more than previously thought. J. Assist. Reprod. Genet. 2014, 31, 627–636. [Google Scholar] [CrossRef] [Green Version]
- Jenkins, T.G.; Aston, K.I.; Meyer, T.D.; Hotaling, J.M.; Shamsi, M.B.; Johnstone, E.B.; Cox, K.J.; Stanford, J.B.; Porucznik, C.A.; Carrell, D.T. Decreased fecundity and sperm DNA methylation patterns. Fertil. Steril. 2016, 105, 51–57.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hartmann, S.; Bergmann, M.; Bohle, R.M.; Weidner, W.; Steger, K. Genetic imprinting during impaired spermatogenesis. Mol. Hum. Reprod. 2006, 12, 407–411. [Google Scholar] [CrossRef]
- Nasri, F.; Gharesi-Fard, B.; Namavar Jahromi, B.; Farazi-Fard, M.A.; Banaei, M.; Davari, M.; Ebrahimi, S.; Anvar, Z. Sperm DNA methylation of H19 imprinted gene and male infertility. Andrologia. 2017, 49. [Google Scholar] [CrossRef]
- Niemitz, E.L.; Feinberg, A.P. Epigenetics and Assisted Reproductive Technology: A Call for Investigation. Am. J. Hum. Genet. 2004, 74, 599–609. [Google Scholar] [CrossRef] [Green Version]
- Tang, Q.; Pan, F.; Yang, J.; Fu, Z.; Lu, Y.; Wu, X.; Han, X.; Chen, M.; Lu, C.; Xia, Y.; et al. Idiopathic male infertility is strongly associated with aberrant DNA methylation of imprinted loci in sperm: A case-control study. Clin. Epigenetics 2018, 10, 134. [Google Scholar] [CrossRef]
- Santi, D.; De Vincentis, S.; Magnani, E.; Spaggiari, G. Impairment of sperm DNA methylation in male infertility: A meta-analytic study. Andrology 2017, 5, 695–703. [Google Scholar] [CrossRef] [Green Version]
- Luján, S.; Caroppo, E.; Niederberger, C.; Arce, J.C.; Sadler-Riggleman, I.; Beck, D.; Nilsson, E.; Skinner, M.K. Sperm DNA Methylation Epimutation Biomarkers for Male Infertility and FSH Therapeutic Responsiveness. Sci. Rep. 2019, 9, 16786. [Google Scholar] [CrossRef] [Green Version]
- Schneider, S.; Balbach, M.; Jikeli, J.F.; Fietz, D.; Nettersheim, D.; Jostes, S.; Schmidt, R.; Kressin, M.; Bergmann, M.; Wachten, D.; et al. Re-visiting the Protamine-2 locus: Deletion, but not haploinsufficiency, renders male mice infertile. Sci. Rep. 2016, 6, 36764. [Google Scholar] [CrossRef]
- Bench, G.; Corzett, M.H.; De Yebra, L.; Oliva, R.; Balhorn, R. Protein and DNA contents in sperm from an infertile human male possessing protamine defects that vary over time. Mol. Reprod. Dev. 1998, 50, 345–353. [Google Scholar] [CrossRef]
- Azpiazu, R.; Amaral, A.; Castillo, J.; Estanyol, J.M.; Guimerà, M.; Ballescà, J.L.; Balasch, J.; Oliva, R. High-throughput sperm differential proteomics suggests that epigenetic alterations contribute to failed assisted reproduction. Hum. Reprod. 2014, 29, 1225–1237. [Google Scholar] [CrossRef] [Green Version]
- Wang, T.; Gao, H.; Li, W.; Liu, C. Essential Role of Histone Replacement and Modifications in Male Fertility. Front. Genet. 2019, 10. [Google Scholar] [CrossRef]
- Bianchi, E.; Boekelheide, K.; Sigman, M.; Braun, J.M.; Eliot, M.; Hall, S.J.; Dere, E.; Hwang, K. Spermatozoal large RNA content is associated with semen characteristics, sociodemographic and lifestyle factors. PLoS ONE 2019, 14, e0216584. [Google Scholar] [CrossRef] [Green Version]
- Hua, M.; Liu, W.; Chen, Y.; Zhang, F.; Xu, B.; Liu, S.; Chen, G.; Shi, H.; Wu, L. Identification of small non-coding RNAs as sperm quality biomarkers for in vitro fertilization. Cell Discov. 2019, 5, 20. [Google Scholar] [CrossRef] [Green Version]
- Godia, M.; Swanson, G.; Krawetz, S.A. A history of why fathers’ RNA matters†. Biol. Reprod. 2018, 99, 147–159. [Google Scholar] [CrossRef] [Green Version]
- Cheung, S.; Parrella, A.; Rosenwaks, Z.; Palermo, G.D. Genetic and epigenetic profiling of the infertile male. PLoS ONE 2019, 14, e0214275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peretz, J.; Vrooman, L.; Ricke, W.A.; Hunt, P.A.; Ehrlich, S.; Hauser, R.; Padmanabhan, V.; Taylor, H.S.; Swan, S.H.; VandeVoort, C.A.; et al. Bisphenol A and Reproductive Health: Update of Experimental and Human Evidence, 2007–2013. Environ. Health Perspect. 2014, 122, 775–786. [Google Scholar] [CrossRef]
- Tavares, R.S.; Escada-Rebelo, S.; Correia, M.; Mota, P.C.; Ramalho-Santos, J. The non-genomic effects of endocrine-disrupting chemicals on mammalian sperm. Reproduction 2016, 151, R1–R13. [Google Scholar] [CrossRef] [Green Version]
- Clark, M.; Hill, J.; Tilman, D. The Diet, Health, and Environment Trilemma. Annu. Rev. Environ. Resour. 2018, 43, 109–134. [Google Scholar] [CrossRef] [Green Version]
- Tiffon, C. The Impact of Nutrition and Environmental Epigenetics on Human Health and Disease. Int. J. Mol. Sci. 2018, 19, 3425. [Google Scholar] [CrossRef] [Green Version]
- Reamon-Buettner, S.M.; Mutschler, V.; Borlak, J. The next innovation cycle in toxicogenomics: Environmental epigenetics. Mutat. Res.-Rev. Mutat. Res. 2008, 659, 158–165. [Google Scholar] [CrossRef]
- Ryberg, K.R.; Gilliom, R.J. Trends in pesticide concentrations and use for major rivers of the United States. Sci. Total Environ. 2015, 538, 431–444. [Google Scholar] [CrossRef]
- Suzawa, M.; Ingraham, H.A. The Herbicide Atrazine Activates Endocrine Gene Networks via Non-Steroidal NR5A Nuclear Receptors in Fish and Mammalian Cells. PLoS ONE 2008, 3, e2117. [Google Scholar] [CrossRef]
- Davis, A.P.; Grondin, C.J.; Lennon-Hopkins, K.; Saraceni-Richards, C.; Sciaky, D.; King, B.L.; Wiegers, T.C.; Mattingly, C.J. The Comparative Toxicogenomics Database’s 10th year anniversary: Update 2015. Nucleic Acids Res. 2015, 43, D914–D920. [Google Scholar] [CrossRef] [Green Version]
- Pogrmic-Majkic, K.; Fa, S.; Dakic, V.; Kaisarevic, S.; Kovacevic, R. upregulation of peripubertal rat leydig cell steroidogenesis following 24 h in vitro and in vivo exposure to atrazine. Toxicol. Sci. 2010, 118, 52–60. [Google Scholar] [CrossRef] [Green Version]
- Victor-Costa, A.B.; Bandeira, S.M.C.; Oliveira, A.G.; Mahecha, G.A.B.; Oliveira, C.A. Changes in testicular morphology and steroidogenesis in adult rats exposed to Atrazine. Reprod. Toxicol. 2010, 29, 323–331. [Google Scholar] [CrossRef]
- Gely-Pernot, A.; Hao, C.; Becker, E.; Stuparevic, I.; Kervarrec, C.; Chalmel, F.; Primig, M.; Jégou, B.; Smagulova, F. The epigenetic processes of meiosis in male mice are broadly affected by the widely used herbicide atrazine. BMC Genomics 2015, 16, 885. [Google Scholar] [CrossRef]
- Cooper, R.L.; Stoker, T.E.; Goldman, J.M.; Parrish, M.B.; Tyrey, L. Effect of atrazine on ovarian function in the rat. Reprod. Toxicol. 1996, 10, 257–264. [Google Scholar] [CrossRef]
- Juliani, C.C.; Silva-Zacarin, E.C.M.; Santos, D.C.; Boer, P.A. Effects of atrazine on female Wistar rats: Morphological alterations in ovarian follicles and immunocytochemical labeling of 90kDa heat shock protein. Micron 2008, 39, 607–616. [Google Scholar] [CrossRef]
- Samardzija, D.; Pogrmic-Majkic, K.; Fa, S.; Glisic, B.; Stanic, B.; Andric, N. Atrazine blocks ovulation via suppression of Lhr and Cyp19a1 mRNA and estradiol secretion in immature gonadotropin-treated rats. Reprod. Toxicol. 2016, 61, 10–18. [Google Scholar] [CrossRef] [PubMed]
- Bardullas, U.; Giordano, M.; Rodríguez, V.M. Chronic atrazine exposure causes disruption of the spontaneous locomotor activity and alters the striatal dopaminergic system of the male Sprague–Dawley rat. Neurotoxicol. Teratol. 2011, 33, 263–272. [Google Scholar] [CrossRef] [PubMed]
- Gojmerac, T.; Kartal, B.; Žurić, M.; Ćurić, S.; Mitak, M. Serum biochemical and histopathological changes related to the hepatic function in pigs following atrazine treatment. J. Appl. Toxicol. 1995, 15, 233–236. [Google Scholar] [CrossRef] [PubMed]
- Islam, M.O.; Hara, M.; Miyake, J. Induction of P-glycoprotein, glutathione-S-transferase and cytochrome P450 in rat liver by atrazine. Environ. Toxicol. Pharmacol. 2002, 12, 1–6. [Google Scholar] [CrossRef]
- Jin, Y.; Lin, X.; Miao, W.; Wang, L.; Wu, Y.; Fu, Z. Oral exposure of pubertal male mice to endocrine-disrupting chemicals alters fat metabolism in adult livers. Environ. Toxicol. 2015, 30, 1434–1444. [Google Scholar] [CrossRef]
- Chevrier, C.; Limon, G.; Monfort, C.; Rouget, F.; Garlantézec, R.; Petit, C.; Durand, G.; Cordier, S. Urinary Biomarkers of Prenatal Atrazine Exposure and Adverse Birth Outcomes in the PELAGIE Birth Cohort. Environ. Health Perspect. 2011, 119, 1034–1041. [Google Scholar] [CrossRef]
- Hao, C.; Gely-Pernot, A.; Kervarrec, C.; Boudjema, M.; Becker, E.; Khil, P.; Tevosian, S.; Jégou, B.; Smagulova, F. 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 Res. 2016, 44, 9784–9802. [Google Scholar] [CrossRef] [Green Version]
- Kelce, W.R.; Stone, C.R.; Laws, S.C.; Gray, L.E.; Kemppainen, J.A.; Wilson, E.M. Persistent DDT metabolite p,p’–DDE is a potent androgen receptor antagonist. Nature 1995, 375, 581–585. [Google Scholar] [CrossRef]
- Song, Y.; Wu, N.; Wang, S.; Gao, M.; Song, P.; Lou, J.; Tan, Y.; Liu, K. Transgenerational impaired male fertility with an Igf2 epigenetic defect in the rat are induced by the endocrine disruptor p, p′-DDE. Hum. Reprod. 2014, 29, 2512–2521. [Google Scholar] [CrossRef] [Green Version]
- Shi, Y.Q.; Wang, Y.P.; Song, Y.; Li, H.W.; Liu, C.J.; Wu, Z.G.; Yang, K.-D. p,p′-DDE induces testicular apoptosis in prepubertal rats via the Fas/FasL pathway. Toxicol. Lett. 2010, 193, 79–85. [Google Scholar] [CrossRef]
- Shi, Y.-Q.; Li, H.-W.; Wang, Y.-P.; Liu, C.-J.; Yang, K.-D. p,p’-DDE induces apoptosis and mRNA expression of apoptosis-associated genes in testes of pubertal rats. Environ. Toxicol. 2013, 28, 31–41. [Google Scholar] [CrossRef]
- Guerrero-Bosagna, C.; Savenkova, M.; Haque, M.M.; Nilsson, E.; Skinner, M.K. Environmentally induced epigenetic transgenerational inheritance of altered Sertoli cell transcriptome and epigenome: Molecular etiology of male infertility. PLoS ONE 2013, 8, e59922. [Google Scholar] [CrossRef]
- Grootegoed, J.A.; Jansen, R.; Van Der Molen, H.J. The role of glucose, pyruvate and lactate in ATP production by rat spermatocytes and spermatids. Biochim. Biophys. Acta (BBA)-Bioenerg. 1984, 767, 248–256. [Google Scholar] [CrossRef]
- Rato, L.; Alves, M.G.; Socorro, S.; Duarte, A.I.; Cavaco, J.E.; Oliveira, P.F. Metabolic regulation is important for spermatogenesis. Nat. Rev. Urol. 2012, 9, 330–338. [Google Scholar] [CrossRef] [PubMed]
- Priya, E.S.; Kumar, T.S.; Singh, P.R.; Balakrishnan, S.; Arunakaran, J. Impact of Lactational Exposure to Polychlorinated Biphenyl Causes Epigenetic Modification and Impairs Sertoli Cells Functional Regulators in F 1 Progeny. Reprod. Sci. 2018, 25, 818–829. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Zhang, P.; Zhao, Y.; Zhang, H. Low dose carbendazim disrupts mouse spermatogenesis might be through estrogen receptor related histone and DNA methylation. Ecotoxicol. Environ. Saf. 2019, 176, 242–249. [Google Scholar] [CrossRef] [PubMed]
- Zhang, P.; Zhao, Y.; Zhang, H.; Liu, J.; Feng, Y.; Yin, S.; Cheng, S.; Sun, X.; Min, L.; Li, L.; et al. Low dose chlorothalonil impairs mouse spermatogenesis through the intertwining of Estrogen Receptor Pathways with histone and DNA methylation. Chemosphere 2019, 230, 384–395. [Google Scholar] [CrossRef]
- Li, H.; Zhang, P.; Zhao, Y.; Zhang, H. Low doses of carbendazim and chlorothalonil synergized to impair mouse spermatogenesis through epigenetic pathways. Ecotoxicol. Environ. Saf. 2020, 188, 109908. [Google Scholar] [CrossRef]
- Santoro, A.; Chianese, R.; Troisi, J.; Richards, S.; Nori, S.L.; Fasano, S.; Guida, M.; Plunk, E.; Viggiano, A.; Pierantoni, R.; et al. Neuro-toxic and reproductive effects of BPA. Curr. Neuropharmacol. 2019, 17, 1109–1132. [Google Scholar] [CrossRef]
- Chianese, R.; Troisi, J.; Richards, S.; Scafuro, M.; Fasano, S.; Guida, M.; Pierantoni, R.; Meccariello, R. Bisphenol A in Reproduction: Epigenetic Effects. Curr. Med. Chem. 2018, 25, 748–770. [Google Scholar] [CrossRef]
- Brotons, J.A.; Olea-Serrano, M.F.; Villalobos, M.; Pedraza, V.; Olea, N. Xenoestrogens released from lacquer coatings in food cans. Environ. Health Perspect. 1995, 103, 608–612. [Google Scholar] [CrossRef]
- Ouchi, K.; Watanabe, S. Measurement of bisphenol A in human urine using liquid chromatography with multi-channel coulometric electrochemical detection. J. Chromatogr. B 2002, 780, 365–370. [Google Scholar] [CrossRef]
- Doshi, T.; Mehta, S.S.; Dighe, V.; Balasinor, N.; Vanage, G. Hypermethylation of estrogen receptor promoter region in adult testis of rats exposed neonatally to bisphenol A. Toxicology 2011, 289, 74–82. [Google Scholar] [CrossRef] [PubMed]
- Salian, S.; Doshi, T.; Vanage, G. Impairment in protein expression profile of testicular steroid receptor coregulators in male rat offspring perinatally exposed to Bisphenol A. Life Sci. 2009, 85, 11–18. [Google Scholar] [CrossRef] [PubMed]
- Yin, L.; Dai, Y.; Jiang, X.; Liu, Y.; Chen, H.; Han, F.; Cao, J.; Liu, J. Role of DNA methylation in bisphenol A exposed mouse spermatocyte. Environ. Toxicol. Pharmacol. 2016, 48, 265–271. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Duan, F.; Zhou, X.; Pan, H.; Li, R. Differential responses of GC-1 spermatogonia cells to high and low doses of bisphenol A. Mol. Med. Rep. 2018, 18, 3034–3040. [Google Scholar] [CrossRef]
- Chen, Z.; Zuo, X.; He, D.; Ding, S.; Xu, F.; Yang, H.; Jin, X.; Fan, Y.; Ying, L.; Tian, C.; et al. Long-term exposure to a ‘safe’ dose of bisphenol A reduced protein acetylation in adult rat testes. Sci. Rep. 2017, 7, 40337. [Google Scholar] [CrossRef]
- Chianese, R.; Viggiano, A.; Urbanek, K.; Cappetta, D.; Troisi, J.; Scafuro, M.; Guida, M.; Esposito, G.; Ciuffreda, L.P.; Rossi, F.; et al. Chronic exposure to low dose of bisphenol A impacts on the first round of spermatogenesis via SIRT1 modulation. Sci. Rep. 2018. [Google Scholar] [CrossRef] [Green Version]
- Cho, H.; Kim, S.J.; Park, H.W.; Oh, M.J.; Yu, S.Y.; Lee, S.Y.; Park, C.; Han, J.R.; Oh, J.H.; Hwang, S.Y.; et al. A relationship between miRNA and gene expression in the mouse Sertoli cell line after exposure to bisphenol A. BioChip J. 2010, 4, 75–81. [Google Scholar] [CrossRef]
- Quintanilla-Vega, B.; Hoover, D.J.; Bal, W.; Silbergeld, E.E.; Waalkes, M.P.; Anderson, L.D. Lead interaction with human protamine (HP2) as a mechanism of male reproductive toxicity. Chem. Res. Toxicol. 2000, 13, 594–600. [Google Scholar] [CrossRef]
- Gao, F.; Zhang, P.; Zhang, H.; Zhang, Y.; Zhang, Y.; Hao, Q.; Zhang, X. Dysregulation of long noncoding RNAs in mouse testes and spermatozoa after exposure to cadmium. Biochem. Biophys. Res. Commun. 2017, 484, 8–14. [Google Scholar] [CrossRef]
- Lv, Y.; Zhang, P.; Guo, J.; Zhu, Z.; Li, X.; Xu, D.; Zeng, W. Melatonin protects mouse spermatogonial stem cells against hexavalent chromium-induced apoptosis and epigenetic histone modification. Toxicol. Appl. Pharmacol. 2018, 340, 30–38. [Google Scholar] [CrossRef] [PubMed]
- Zhang, T.; Zhou, Y.; Li, L.; Zhao, Y.; De Felici, M.; Reiter, R.J.; Shen, W. Melatonin protects prepuberal testis from deleterious effects of bisphenol A or diethylhexyl phthalate by preserving H3K9 methylation. J. Pineal Res. 2018, 65, e12497. [Google Scholar] [CrossRef] [PubMed]
- Prados, J.; Stenz, L.; Somm, E.; Stouder, C.; Dayer, A.; Paoloni-Giacobino, A. Prenatal exposure to DEHP affects spermatogenesis and sperm DNA methylation in a strain-dependent manner. PLoS ONE 2015, 10, e0132136. [Google Scholar] [CrossRef] [PubMed]
- Do, R.P.; Stahlhut, R.W.; Ponzi, D.; vom Saal, F.S.; Taylor, J.A. Non-monotonic dose effects of in utero exposure to di(2-ethylhexyl) phthalate (DEHP) on testicular and serum testosterone and anogenital distance in male mouse fetuses. Reprod. Toxicol. 2012, 34, 614–621. [Google Scholar] [CrossRef] [Green Version]
- Parks, L.G.; Ostby, J.S.; Lambright, C.R.; Abbott, B.D.; Klinefelter, G.R.; Barlow, N.J.; Gray, L.E. The plasticizer diethylhexyl phthalate induces malformations by decreasing fetal testosterone synthesis during sexual differentiation in the male rat. Toxicol. Sci. 2000, 58, 339–349. [Google Scholar] [CrossRef]
- Swan, S.H.; Main, K.M.; Liu, F.; Stewart, S.L.; Kruse, R.L.; Calafat, A.M.; Mao, C.S.; Redmon, J.B.; Ternand, C.L.; Sullivan, S.; et al. Decrease in anogenital distance among male infants with prenatal phthalate exposure. Environ. Health Perspect. 2005, 113, 1056–1061. [Google Scholar] [CrossRef] [Green Version]
- Doyle, T.J.; Bowman, J.L.; Windell, V.L.; McLean, D.J.; Kim, K.H. Transgenerational Effects of Di-(2-ethylhexyl) Phthalate on Testicular Germ Cell Associations and Spermatogonial Stem Cells in Mice1. Biol. Reprod. 2013, 88, 112. [Google Scholar] [CrossRef]
- Pang, J.; Zhou, Q.; Sun, X.; Li, L.; Zhou, B.; Zeng, F.; Zhao, Y.; Shen, W.; Sun, Z. Effect of low-dose zearalenone exposure on reproductive capacity of male mice. Toxicol. Appl. Pharmacol. 2017, 333, 60–67. [Google Scholar] [CrossRef]
- Gao, Y.; Zhao, Y.; Zhang, H.; Zhang, P.; Liu, J.; Feng, Y.; Men, Y.; Li, L.; Shen, W.; Sun, Z.; et al. Pubertal exposure to low doses of zearalenone disrupting spermatogenesis through ERα related genetic and epigenetic pathways. Toxicol. Lett. 2019, 315, 31–38. [Google Scholar] [CrossRef]
- Men, Y.; Zhao, Y.; Zhang, P.; Zhang, H.; Gao, Y.; Liu, J.; Feng, Y.; Li, L.; Shen, W.; Sun, Z.; et al. Gestational exposure to low-dose zearalenone disrupting offspring spermatogenesis might be through epigenetic modifications. Basic Clin. Pharmacol. Toxicol. 2019, 125, 382–393. [Google Scholar] [CrossRef]
- Meccariello, R.; Chianese, R.; Ciaramella, V.; Fasano, S.; Pierantoni, R. Molecular Chaperones, Cochaperones, and Ubiquitination/Deubiquitination System: Involvement in the Production of High Quality Spermatozoa. Biomed Res. Int. 2014, 2014, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Meccariello, R.; Chianese, R.; Chioccarelli, T.; Ciaramella, V.; Fasano, S.; Pierantoni, R.; Cobellis, G. Intra-Testicular Signals Regulate Germ Cell Progression and Production of Qualitatively Mature Spermatozoa in Vertebrates. Front. Endocrinol. 2014, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chianese, R.; Cobellis, G.; Chioccarelli, T.; Ciaramella, V.; Migliaccio, M.; Fasano, S.; Pierantoni, R.; Meccariello, R. Kisspeptins, Estrogens and Male Fertility. Curr. Med. Chem. 2016, 23, 4070–4091. [Google Scholar] [CrossRef] [PubMed]
- Younglai, E.V.; Foster, W.G.; Hughes, E.G.; Trim, K.; Jarrell, J.F. Levels of Environmental Contaminants in Human Follicular Fluid, Serum, and Seminal Plasma of Couples Undergoing In Vitro Fertilization. Arch. Environ. Contam. Toxicol. 2002, 43, 121–126. [Google Scholar] [CrossRef] [PubMed]
- Amaral, A.; Lourenço, B.; Marques, M.; Ramalho-Santos, J. Mitochondria functionality and sperm quality. Reproduction 2013, 146, R163–R174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salian, S.; Doshi, T.; Vanage, G. Neonatal exposure of male rats to Bisphenol A impairs fertility and expression of sertoli cell junctional proteins in the testis. Toxicology 2009, 265, 56–67. [Google Scholar] [CrossRef]
- Brouard, V.; Guénon, I.; Bouraima-Lelong, H.; Delalande, C. Differential effects of bisphenol A and estradiol on rat spermatogenesis’ establishment. Reprod. Toxicol. 2016, 63, 49–61. [Google Scholar] [CrossRef]
- Auger, J.; Lesaffre, C.; Bazire, A.; Schoevaert-Brossault, D.; Eustache, F. High-resolution image cytometry of rat sperm nuclear shape, size and chromatin status: Experimental validation with the reproductive toxicant vinclozolin. Reprod. Toxicol. 2004, 18, 775–783. [Google Scholar] [CrossRef]
- Othman, A.I.; Edrees, G.M.; El-Missiry, M.A.; Ali, D.A.; Aboel-Nour, M.; Dabdoub, B.R. Melatonin controlled apoptosis and protected the testes and sperm quality against bisphenol A-induced oxidative toxicity. Toxicol. Ind. Health 2016, 32, 1537–1549. [Google Scholar] [CrossRef]
- De Jager, C. Reduced Seminal Parameters Associated With Environmental DDT Exposure and p,p’-DDE Concentrations in Men in Chiapas, Mexico: A Cross-Sectional Study. J. Androl. 2006, 27, 16–27. [Google Scholar] [CrossRef]
- Siddeek, B.; Lakhdari, N.; Inoubli, L.; Paul-Bellon, R.; Isnard, V.; Thibault, E.; Bongain, A.; Chevallier, D.; Repetto, E.; Trabucchi, M.; et al. Developmental epigenetic programming of adult germ cell death disease: Polycomb protein EZH2–miR-101 pathway. Epigenomics 2016, 8, 1459–1479. [Google Scholar] [CrossRef] [PubMed]
- Lombó, M.; Fernández-Díez, C.; González-Rojo, S.; Herráez, M.P. Genetic and epigenetic alterations induced by bisphenol A exposure during different periods of spermatogenesis: From spermatozoa to the progeny. Sci. Rep. 2019, 9, 18029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, B.; Qi, Y.; Liu, D.; Gao, X.; Chen, H.; Bai, C.; Huang, Z. Cigarette smoking is associated with abnormal histone-to-protamine transition in human sperm. Fertil. Steril. 2014, 101, 51–57.e1. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.K.; Jee, B.C.; Kim, S.H. Histone methylation and acetylation in ejaculated human sperm: Effects of swim-up and smoking. Fertil. Steril. 2015, 103, 1425–1431. [Google Scholar] [CrossRef]
- Dong, H.; Wang, Y.; Zou, Z.; Chen, L.; Shen, C.; Xu, S.; Zhang, J.; Zhao, F.; Ge, S.; Gao, Q.; et al. Abnormal methylation of imprinted genes and cigarette smoking: Assessment of their association with the risk of male infertility. Reprod. Sci. 2017, 24, 114–123. [Google Scholar] [CrossRef]
- Bell, E.L.; Nagamori, I.; Williams, E.O.; Del Rosario, A.M.; Bryson, B.D.; Watson, N.; White, F.M.; Sassone-Corsi, P.; Guarente, L. SirT1 is required in the male germ cell for differentiation and fecundity in mice. Development 2014, 141, 3495–3504. [Google Scholar] [CrossRef] [Green Version]
- 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. 2012, 34, 694–707. [Google Scholar] [CrossRef] [Green Version]
- Stouder, C.; Paoloni-Giacobino, A. Specific transgenerational imprinting effects of the endocrine disruptor methoxychlor on male gametes. Reproduction 2011, 141, 207–216. [Google Scholar] [CrossRef] [Green Version]
- Miao, M.; Zhou, X.; Li, Y.; Zhang, O.; Zhou, Z.; Li, T.; Yuan, W.; Li, R.; Li, D.K. LINE-1 hypomethylation in spermatozoa is associated with Bisphenol A exposure. Andrology 2014, 2, 138–144. [Google Scholar] [CrossRef]
- Meunier, L.; Siddeek, B.; Vega, A.; Lakhdari, N.; Inoubli, L.; Bellon, R.P.; Lemaire, G.; Mauduit, C.; Benahmed, M. Perinatal programming of adult rat germ cell death after exposure to xenoestrogens: Role of microRNA miR-29 family in the down-regulation of DNA methyltransferases and Mcl-1. Endocrinology 2012, 153, 1936–1947. [Google Scholar] [CrossRef] [Green Version]
- Yauk, C.; Polyzos, A.; Rowan-Carroll, A.; Somers, C.M.; Godschalk, R.W.; Van Schooten, F.J.; Berndt, M.L.; Pogribny, I.P.; Koturbash, I.; Williams, A.; et al. Germ-line mutations, DNA damage, and global hypermethylation in mice exposed to particulate air pollution in an urban/industrial location. Proc. Natl. Acad. Sci. USA 2008, 105, 605–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shiao, Y.H.; Crawford, E.B.; Anderson, L.M.; Patel, P.; Ko, K. Allele-specific germ cell epimutation in the spacer promoter of the 45S ribosomal RNA gene after Cr(III) exposure. Toxicol. Appl. Pharmacol. 2005, 205, 290–296. [Google Scholar] [CrossRef] [PubMed]
- Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Skinner, M.K. Dioxin (TCDD) Induces Epigenetic Transgenerational Inheritance of Adult Onset Disease and Sperm Epimutations. PLoS ONE 2012, 7, e46249. [Google Scholar] [CrossRef] [PubMed]
- Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Skinner, M.K. Plastics Derived Endocrine Disruptors (BPA, DEHP and DBP) Induce Epigenetic Transgenerational Inheritance of Obesity, Reproductive Disease and Sperm Epimutations. PLoS ONE 2013, 8, e55387. [Google Scholar] [CrossRef]
- Anway, M.D.; Memon, M.A.; Uzumcu, M.; Skinner, M.K. Transgenerational Effect of the Endocrine Disruptor Vinclozolin on Male Spermatogenesis. J. Androl. 2006, 27, 868–879. [Google Scholar] [CrossRef] [Green Version]
- Anway, M.D.; Leathers, C.; Skinner, M.K. Endocrine disruptor vinclozolin induced epigenetic transgenerational adult-onset disease. Endocrinology 2006, 6, 868–879. [Google Scholar] [CrossRef] [Green Version]
- Guerrero-Bosagna, C.; Settles, M.; Lucker, B.; Skinner, M.K. Epigenetic Transgenerational Actions of Vinclozolin on Promoter Regions of the Sperm Epigenome. PLoS ONE 2010, 5, e13100. [Google Scholar] [CrossRef] [Green Version]
- Nilsson, E.; King, S.E.; McBirney, M.; Kubsad, D.; Pappalardo, M.; Beck, D.; Sadler-Riggleman, I.; Skinner, M.K. Vinclozolin induced epigenetic transgenerational inheritance of pathologies and sperm epimutation biomarkers for specific diseases. PLoS ONE 2018, 13, e0202662. [Google Scholar] [CrossRef]
- Manikkam, M.; Guerrero-Bosagna, C.; Tracey, R.; Haque, M.M.; Skinner, M.K. Transgenerational actions of environmental compounds on reproductive disease and identification of epigenetic biomarkers of ancestral exposures. PLoS ONE 2012, 7, e31901. [Google Scholar] [CrossRef] [Green Version]
- Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Skinner, M.K. Pesticide and insect repellent mixture (permethrin and DEET) induces epigenetic transgenerational inheritance of disease and sperm epimutations. Reprod. Toxicol. 2012, 34, 708–719. [Google Scholar] [CrossRef] [Green Version]
- Tracey, R.; Manikkam, M.; Guerrero-Bosagna, C.; Skinner, M.K. Hydrocarbons (jet fuel JP-8) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations. Reprod. Toxicol. 2013, 36, 104–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kubsad, D.; Nilsson, E.E.; King, S.E.; Sadler-Riggleman, I.; Beck, D.; Skinner, M.K. Assessment of Glyphosate Induced Epigenetic Transgenerational Inheritance of Pathologies and Sperm Epimutations: Generational Toxicology. Sci. Rep. 2019, 9, 6372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skinner, M.K.; Manikkam, M.; Tracey, R.; Guerrero-Bosagna, C.; Haque, M.; Nilsson, E.E. Ancestral dichlorodiphenyltrichloroethane (DDT) exposure promotes epigenetic transgenerational inheritance of obesity. BMC Med. 2013, 11, 228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, Y.; Yang, L. Transgenerational impaired spermatogenesis with sperm H19 and Gtl2 hypomethylation induced by the endocrine disruptor p,p’-DDE. Toxicol. Lett. 2018, 297, 34–41. [Google Scholar] [CrossRef]
- King, S.E.; McBirney, M.; Beck, D.; Sadler-Riggleman, I.; Nilsson, E.; Skinner, M.K. Sperm epimutation biomarkers of obesity and pathologies following DDT induced epigenetic transgenerational inheritance of disease. Environ. Epigenetics 2019, 5, dvz008. [Google Scholar] [CrossRef] [Green Version]
- Ben Maamar, M.; Nilsson, E.; Sadler-Riggleman, I.; Beck, D.; McCarrey, J.R.; Skinner, M.K. Developmental origins of transgenerational sperm DNA methylation epimutations following ancestral DDT exposure. Dev. Biol. 2019, 445, 280–293. [Google Scholar] [CrossRef]
- Skinner, M.K.; Nilsson, E.; Sadler-Riggleman, I.; Beck, D.; Ben Maamar, M.; McCarrey, J.R. Transgenerational sperm DNA methylation epimutation developmental origins following ancestral vinclozolin exposure. Epigenetics 2019, 14, 721–739. [Google Scholar] [CrossRef] [Green Version]
- Schneider, S.; Kaufmann, W.; Buesen, R.; van Ravenzwaay, B. Vinclozolin-The lack of a transgenerational effect after oral maternal exposure during organogenesis. Reprod. Toxicol. 2008, 25, 352–360. [Google Scholar] [CrossRef]
- Inawaka, K.; Kawabe, M.; Takahashi, S.; Doi, Y.; Tomigahara, Y.; Tarui, H.; Abe, J.; Kawamura, S.; Shirai, T. Maternal exposure to anti-androgenic compounds, vinclozolin, flutamide and procymidone, has no effects on spermatogenesis and DNA methylation in male rats of subsequent generations. Toxicol. Appl. Pharmacol. 2009, 237, 178–187. [Google Scholar] [CrossRef]
- Stouder, C.; Paoloni-Giacobino, A. Transgenerational effects of the endocrine disruptor vinclozolin on the methylation pattern of imprinted genes in the mouse sperm. Reproduction 2010, 139, 373–379. [Google Scholar] [CrossRef] [Green Version]
- Brieño-Enríquez, M.A.; García-López, J.; Cárdenas, D.B.; Guibert, S.; Cleroux, E.; Děd, L.; Hourcade, J.D.D.; Pěknicová, J.; Weber, M.; del Mazo, J. Exposure to endocrine disruptor induces transgenerational epigenetic deregulation of microRNAs in primordial germ cells. PLoS ONE 2015, 10, e0124296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, T.; Mokshagundam, S.; Rinaudo, P.F.; Osteen, K.G.; Bruner-Tran, K.L. Paternal developmental toxicant exposure is associated with epigenetic modulation of sperm and placental Pgr and Igf2 in a mouse model. Biol. Reprod. 2018, 99, 864–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Filkowski, J.N.; Ilnytskyy, Y.; Tamminga, J.; Koturbash, I.; Golubov, A.; Bagnyukova, T.; Pogribny, I.P.; Kovalchuk, O. Hypomethylation and genome instability in the germline of exposed parents and their progeny is associated with altered miRNA expression. Carcinogenesis 2010, 31, 1110–1115. [Google Scholar] [CrossRef] [PubMed]
- Gapp, K.; Jawaid, A.; Sarkies, P.; Bohacek, J.; Pelczar, P.; Prados, J.; Farinelli, L.; Miska, E.; Mansuy, I.M. Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nat. Neurosci. 2014. [Google Scholar] [CrossRef] [Green Version]
- McPherson, N.O.; Owens, J.A.; Fullston, T.; Lane, M. Preconception diet or exercise intervention in obese fathers normalizes sperm microRNA profile and metabolic syndrome in female offspring. Am. J. Physiol. Metab. 2015, 308, E805–E821. [Google Scholar] [CrossRef] [Green Version]
- Ashe, A.; Sapetschnig, A.; Weick, E.M.; Mitchell, J.; Bagijn, M.P.; Cording, A.C.; Doebley, A.L.; Goldstein, L.D.; Lehrbach, N.J.; Le Pen, J.; et al. PiRNAs can trigger a multigenerational epigenetic memory in the germline of C. elegans. Cell 2012, 150, 88–99. [Google Scholar] [CrossRef] [Green Version]
- Ben Maamar, M.; Sadler-Riggleman, I.; Beck, D.; McBirney, M.; Nilsson, E.; Klukovich, R.; Xie, Y.; Tang, C.; Yan, W.; Skinner, M.K. Alterations in sperm DNA methylation, non-coding RNA expression, and histone retention mediate vinclozolin-induced epigenetic transgenerational inheritance of disease. Environ. Epigenetics 2018, 4. [Google Scholar] [CrossRef]
- Shi, M.; Whorton, A.E.; Sekulovski, N.; MacLean, J.A.; Hayashi, K. Prenatal Exposure to Bisphenol A, E, and S Induces Transgenerational Effects on Male Reproductive Functions in Mice. Toxicol. Sci. 2019, 172, 303–315. [Google Scholar] [CrossRef]
- Hughes, V. Epigenetics: The sins of the father. Nature 2014, 507, 22–24. [Google Scholar] [CrossRef]
- Li, Y. Epigenetic mechanisms link maternal diets and gut microbiome to obesity in the offspring. Front. Genet. 2018, 9, 1–13. [Google Scholar] [CrossRef]
- Grumati, P.; Coletto, L.; Sabatelli, P.; Cescon, M.; Angelin, A.; Bertaggia, E.; Blaauw, B.; Urciuolo, A.; Tiepolo, T.; Merlini, L.; et al. Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration. Nat. Med. 2010, 16, 1313–1320. [Google Scholar] [CrossRef] [PubMed]
- Boccardi, V.; Paolisso, G.; Mecocci, P. Nutrition and lifestyle in healthy aging: The telomerase challenge. Aging 2016, 8, 12–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castagnaro, S.; Pellegrini, C.; Pellegrini, M.; Chrisam, M.; Sabatelli, P.; Toni, S.; Grumati, P.; Ripamonti, C.; Pratelli, L.; Maraldi, N.M.; et al. Autophagy activation in COL6 myopathic patients by a low-protein-diet pilot trial. Autophagy 2016, 12, 2484–2495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camandola, S.; Mattson, M.P. Brain metabolism in health, aging, and neurodegeneration. EMBO J. 2017, 36, 1474–1492. [Google Scholar] [CrossRef] [PubMed]
- Schagdarsurengin, U.; Steger, K. Epigenetics in male reproduction: Effect of paternal diet on sperm quality and offspring health. Nat. Rev. Urol. 2016, 13, 584–595. [Google Scholar] [CrossRef] [PubMed]
- Ng, S.F.; Lin, R.C.Y.; Laybutt, D.R.; Barres, R.; Owens, J.A.; Morris, M.J. Chronic high-fat diet in fathers programs β 2-cell dysfunction in female rat offspring. Nature 2010, 467, 963–966. [Google Scholar] [CrossRef]
- Fullston, T.; Teague, E.M.C.O.; Palmer, N.O.; Deblasio, M.J.; Mitchell, M.; Corbett, M.; Print, C.G.; Owens, J.A.; Lane, M. Paternal obesity initiates metabolic disturbances in two generations of mice with incomplete penetrance to the F2 generation and alters the transcriptional profile of testis and sperm microRNA content. FASEB J. 2013, 27, 4226–4243. [Google Scholar] [CrossRef]
- Soubry, A. Epigenetic inheritance and evolution: A paternal perspective on dietary influences. Prog. Biophys. Mol. Biol. 2015, 118, 79–85. [Google Scholar] [CrossRef] [Green Version]
- De Castro Barbosa, T.; Ingerslev, L.R.; Alm, P.S.; Versteyhe, S.; Massart, J.; Rasmussen, M.; Donkin, I.; Sjögren, R.; Mudry, J.M.; Vetterli, L.; et al. High-fat diet reprograms the epigenome of rat spermatozoa and transgenerationally affects metabolism of the offspring. Mol. Metab. 2016, 5, 184–197. [Google Scholar] [CrossRef]
- 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. USA 2014, 111, 1873–1878. [Google Scholar] [CrossRef] [Green Version]
- Carone, B.R.; Fauquier, L.; Habib, N.; Shea, J.M.; Hart, C.E.; Li, R.; Bock, C.; Li, C.; Gu, H.; Zamore, P.D.; et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 2010, 143, 1084–1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Siklenka, K.; Erkek, S.; Godmann, M.; Lambrot, R.; McGraw, S.; Lafleur, C.; Cohen, T.; Xia, J.; Suderman, M.; Hallett, M.; et al. Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science 2015, 350, aab2006. [Google Scholar] [CrossRef] [PubMed]
- Terashima, M.; Barbour, S.; Ren, J.; Yu, W.; Han, Y.; Muegge, K. Effect of high fat diet on paternal sperm histone distribution and male offspring liver gene expression. Epigenetics 2015, 10, 861–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palmer, N.O.; Fullston, T.; Mitchell, M.; Setchell, B.P.; Lane, M. SIRT6 in mouse spermatogenesis is modulated by diet-induced obesity. Reprod. Fertil. Dev. 2011, 23, 929–939. [Google Scholar] [CrossRef] [PubMed]
- Houfflyn, S.; Matthys, C.; Soubry, A. Correction to: Male obesity: Epigenetic origin and effects in sperm and offspring. Curr. Mol. Biol. Rep. 2018, 4, 24. [Google Scholar] [CrossRef] [Green Version]
- Holoch, D.; Moazed, D. RNA-mediated epigenetic regulation of gene expression. Nat. Rev. Genet. 2015, 16, 71–84. [Google Scholar] [CrossRef]
- Sharma, U.; Conine, C.C.; Shea, J.M.; Boskovic, A.; Derr, A.G.; Bing, X.Y.; Belleannee, C.; Kucukural, A.; Serra, R.W.; Sun, F.; et al. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science 2016, 351, 391–396. [Google Scholar] [CrossRef] [Green Version]
- Lau, J.; Herzog, H. CART in the regulation of appetite and energy homeostasis. Front. Neurosci. 2014, 8. [Google Scholar] [CrossRef] [Green Version]
- Ingerslev, L.R.; Donkin, I.; Fabre, O.; Versteyhe, S.; Mechta, M.; Pattamaprapanont, P.; Mortensen, B.; Krarup, N.T.; Barrès, R. Endurance training remodels sperm-borne small RNA expression and methylation at neurological gene hotspots. Clin. Epigenetics 2018, 10, 12. [Google Scholar] [CrossRef] [Green Version]
- Radford, E.J.; Ito, M.; Shi, H.; Corish, J.A.; Yamazawa, K.; Isganaitis, E.; Seisenberger, S.; Hore, T.A.; Reik, W.; Erkek, S.; et al. In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science 2014, 345, 1255903. [Google Scholar] [CrossRef] [Green Version]
- Heijmans, B.T.; Tobi, E.W.; Stein, A.D.; Putter, H.; Blauw, G.J.; Susser, E.S.; Slagboom, P.E.; Lumey, L.H. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc. Natl. Acad. Sci. USA 2008, 105, 17046–17049. [Google Scholar] [CrossRef] [Green Version]
- Watkins, A.J.; Dias, I.; Tsuro, H.; Allen, D.; Emes, R.D.; Moreton, J.; Wilson, R.; Ingram, R.J.M.; Sinclair, K.D. Paternal diet programs offspring health through sperm- and seminal plasma-specific pathways in mice. Proc. Natl. Acad. Sci. USA 2018, 115, 10064–10069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mendiola, J.; Torres-Cantero, A.M.; Vioque, J.; Moreno-Grau, J.M.; Ten, J.; Roca, M.; Moreno-Grau, S.; Bernabeu, R. A low intake of antioxidant nutrients is associated with poor semen quality in patients attending fertility clinics. Fertil. Steril. 2010, 93, 1128–1133. [Google Scholar] [CrossRef] [PubMed]
- Blomberg Jensen, M.; Bjerrum, P.J.; Jessen, T.E.; Nielsen, J.E.; Joensen, U.N.; Olesen, I.A.; Petersen, J.H.; Juul, A.; Dissing, S.; Jørgensen, N. Vitamin D is positively associated with sperm motility and increases intracellular calcium in human spermatozoa. Hum. Reprod. 2011, 26, 1307–1317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pike, J.W.; Meyer, M.B.; Bishop, K.A. Regulation of target gene expression by the vitamin D receptor-an update on mechanisms. Rev. Endocr. Metab. Disord. 2012, 13, 45–55. [Google Scholar] [CrossRef] [PubMed]
- Karlic, H.; Varga, F. Impact of vitamin D metabolism on clinical epigenetics. Clin. Epigenetics 2011, 2, 55–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mejos, K.K.; Kim, H.W.; Lim, E.M.; Chang, N. Effects of parental folate deficiency on the folate content, global DNA methylation, and expressions of FRα, IGF-2 and IGF-1R in the postnatal rat liver. Nutr. Res. Pract. 2013, 7, 281–286. [Google Scholar] [CrossRef]
- 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. 2013, 4, 2889. [Google Scholar] [CrossRef]
- Forges, T.; Monnier-Barbarino, P.; Alberto, J.M.; Guéant-Rodriguez, R.M.; Daval, J.L.; Guéant, J.L. Impact of folate and homocysteine metabolism on human reproductive health. Hum. Reprod. Update 2007, 13, 225–238. [Google Scholar] [CrossRef]
- Wong, W.Y.; Merkus, H.M.W.M.; Thomas, C.M.G.; Menkveld, R.; Zielhuis, G.A.; Steegers-Theunissen, R.P.M. Effects of folic acid and zinc sulfate on male factor subfertility: A double-blind, randomized, placebo-controlled trial. Fertil. Steril. 2002, 77, 491–498. [Google Scholar] [CrossRef]
- Ebisch, I.M.W.; Thomas, C.M.G.; Peters, W.H.M.; Braat, D.D.M.; Steegers-Theunissen, R.P.M. The importance of folate, zinc and antioxidants in the pathogenesis and prevention of subfertility. Hum. Reprod. Update 2007, 13, 163–174. [Google Scholar] [CrossRef] [PubMed]
- Cui, X.; Jing, X.; Wu, X.; Yan, M. Protective effect of resveratrol on spermatozoa function in male infertility induced by excess weight and obesity. Mol. Med. Rep. 2016, 14, 4659–4665. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Diao, R.Y.; Duan, Y.G.; Yi, T.H.; Cai, Z.M. In vitro antioxidant effect of curcumin on human sperm quality in leucocytospermia. Andrologia 2017, 49, e12760. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Song, W.; Wang, Z.; Wang, Z.; Jin, X.; Xu, J.; Bai, L.; Li, Y.; Cui, J.; Cai, L. Resveratrol attenuates testicular apoptosis in type 1 diabetic mice: Role of Akt-mediated Nrf2 activation and p62-dependent Keap1 degradation. Redox Biol. 2018, 14, 609–617. [Google Scholar] [CrossRef] [PubMed]
- Lv, Z.M.; Ling, M.Y.; Chen, C. Comparative proteomics reveals protective effect of resveratrol on a high-fat diet-induced damage to mice testis. Syst. Biol. Reprod. Med. 2020, 66, 37–49. [Google Scholar] [CrossRef] [Green Version]
- Björndahl, L.; Kvist, U. Sequence of ejaculation affects the spermatozoon as a carrier and its message. Reprod. Biomed. Online 2003, 7, 440–448. [Google Scholar] [CrossRef]
- Bjorndahl, L.; Kvist, U. Human sperm chromatin stabilization: A proposed model including zinc bridges. Mol. Hum. Reprod. 2010, 16, 23–29. [Google Scholar] [CrossRef] [Green Version]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cescon, M.; Chianese, R.; Tavares, R.S. Environmental Impact on Male (In)Fertility via Epigenetic Route. J. Clin. Med. 2020, 9, 2520. https://doi.org/10.3390/jcm9082520
Cescon M, Chianese R, Tavares RS. Environmental Impact on Male (In)Fertility via Epigenetic Route. Journal of Clinical Medicine. 2020; 9(8):2520. https://doi.org/10.3390/jcm9082520
Chicago/Turabian StyleCescon, Matilde, Rosanna Chianese, and Renata S. Tavares. 2020. "Environmental Impact on Male (In)Fertility via Epigenetic Route" Journal of Clinical Medicine 9, no. 8: 2520. https://doi.org/10.3390/jcm9082520
APA StyleCescon, M., Chianese, R., & Tavares, R. S. (2020). Environmental Impact on Male (In)Fertility via Epigenetic Route. Journal of Clinical Medicine, 9(8), 2520. https://doi.org/10.3390/jcm9082520