全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Sperm RNA as a Mediator of Genomic Plasticity

DOI: 10.1155/2014/179701

Full-Text   Cite this paper   Add to My Lib

Abstract:

Sperm RNA has been linked recently to trans-generational, non-Mendelian patterns of inheritance. Originally dismissed as “residual” to spermatogenesis, some sperm RNA may have postfertilization functions including the transmission of acquired characteristics. Sperm RNA may help explain how trans-generational effects are transmitted and it may also have implications for assisted reproductive technologies (ART) where sperm are subjected to considerable, ex vivo manual handling. The presence of sperm RNA was originally a controversial topic because nuclear gene expression is switched off in the mature mammalian spermatozoon. With the recent application of next generation sequencing (NGS), an unexpectedly rich and complex repertoire of RNAs has been revealed in the sperm of several species that makes its residual presence counterintuitive. What follows is a personal survey of the science behind our understanding of sperm RNA and its functional significance based on experimental observations from my laboratory as well as many others who have contributed to the field over the years and are continuing to contribute today. The narrative begins with a historical perspective and ends with some educated speculation on where research into sperm RNA is likely to lead us in the next 10 years or so. 1. Introduction The recent publication of two reports on transgenerationally acquired inheritance (henceforth called TAC) in the mouse has brought the subject of sperm RNA back sharply into focus. The first of these [1] examined the transmission of a conditioned aversion to a particular odor in the F0s, which carried through to conditioned (and odor)-na?ve F1 and F2 pups. The second report [2] demonstrated the inheritance of cognitive and behavioural conditioning in the F0s by F1 and F2 pups that had no prior experience of the conditioning (Figure 1). Although only the latter report went on to demonstrate a change in the (small noncoding) sperm RNA profile of conditioned mice that could be linked to the conditioning response and transmission, these reports, in association with earlier reports showing the transmission of paramutation effects by sperm RNA [3], strongly suggest that the odor conditioning was also transmitted by or associated with sperm RNA (or possibly sperm RNA-altered DNA methylation). In hindsight, the connection between sperm RNA and TAC now seems more obvious given that the transmission must go through the germ line [4]. Whether the RNA somehow marks the sperm genome before its entry into the ooplasm at fertilisation or is required to make its mark at

References

[1]  B. G. Dias and K. J. Ressler, “Parental olfactory experience influences behavior and neural structure in subsequent generations,” Nature Neuroscience, vol. 17, no. 1, pp. 89–96, 2014.
[2]  K. Gapp, A. Jawaid, P. Sarkies et al., “Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice,” Nature Neuroscience, vol. 17, no. 5, pp. 667–669, 2014.
[3]  M. Rassoulzadegan, V. Grandjean, P. Gounon, S. Vincent, I. Gillot, and F. Cuzin, “RNA-mediated non-mendelian inheritance of an epigenetic change in the mouse,” Nature, vol. 441, no. 7092, pp. 469–474, 2006.
[4]  M. E. Pembrey, L. O. Bygren, G. Kaati et al., “Sex-specific, male-line transgenerational responses in humans,” European Journal of Human Genetics, vol. 14, no. 2, pp. 159–166, 2006.
[5]  K. Northstone, J. Golding, G. Davey Smith, L. L. Miller, and M. Pembrey, “Prepubertal start of father's smoking and increased body fat in his sons: further characterisation of paternal transgenerational responses,” European Journal of Human Genetics, 2014.
[6]  R. Balhorn, L. Brewer, and M. Corzett, “DNA condensation by protamine and arginine-rich peptides: analysis of toroid stability using single DNA molecules,” Molecular Reproduction and Development, vol. 56, supplement 2, pp. 230–234, 2000.
[7]  G. Fuentes-Mascorro, H. Serrano, and A. Rosado, “Sperm chromatin,” Archives of Andrology, vol. 45, no. 3, pp. 215–225, 2000.
[8]  C. Rathe, W. M. Baarends, S. Jayaramaiah-Raja, M. Bartkuhn, R. Renkawitz, and R. Renkawitz-Pohl, “Transition from a nucleosome-based to a protamine-based chromatin configuration during spermiogenesis in Drosophila,” Journal of Cell Science, vol. 120, part 9, pp. 1689–1700, 2007.
[9]  R. Balhorn, “The protamine family of sperm nuclear proteins,” Genome Biology, vol. 8, no. 9, article 227, 2007.
[10]  E. Rejon, C. Bajon, A. Blaize, and D. Robert, “RNA in the nucleus of a motile plant spermatozoid: characterization by enzyme-gold cytochemistry and in situ hybridization,” Molecular Reproduction and Development, vol. 1, no. 1, pp. 49–56, 1988.
[11]  C. A. Pessot, M. Brito, J. Figueroa, I. I. Concha, A. Yanez, and L. O. Burzio, “Presence of RNA in the sperm nucleus,” Biochemical and Biophysical Research Communications, vol. 158, no. 1, pp. 272–278, 1989.
[12]  S. M. Wykes and S. A. Krawetz, “The structural organization of sperm chromatin,” Journal of Biological Chemistry, vol. 278, no. 32, pp. 29471–29477, 2003.
[13]  W. W. Franke, “Nuclear envelopes: structure and biochemistry of the nuclear envelope,” Philosophical Transactions of the Royal Society of London Series B: Biological Sciences, vol. 268, no. 891, pp. 67–93, 1974.
[14]  G. D. Johnson, C. Lalancette, A. K. Linnemann, F. Leduc, G. Boissonneault, and S. A. Krawetz, “The sperm nucleus: chromatin, RNA, and the nuclear matrix,” Reproduction, vol. 141, no. 1, pp. 21–36, 2011.
[15]  J.-P. Dadoune, “Spermatozoal RNAs: what about their functions?” Microscopy Research and Technique, vol. 72, no. 8, pp. 536–551, 2009.
[16]  M. Jodar, S. Selvaraju, E. Sendler, M. P. Diamond, and S. A. Krawetz, “The presence, role and clinical use of spermatozoal RNAs,” Human Reproduction Update, vol. 19, no. 6, pp. 604–624, 2013.
[17]  C. Lalancette, D. Miller, Y. Li, and S. A. Krawetz, “Paternal contributions: new functional insights for spermatozoal RNA,” Journal of Cellular Biochemistry, vol. 104, no. 5, pp. 1570–1579, 2008.
[18]  D. Miller and G. C. Ostermeier, “Towards a better understanding of RNA carriage by ejaculate spermatozoa,” Human Reproduction Update, vol. 12, no. 6, pp. 757–767, 2006.
[19]  N. B. Hecht, “Molecular mechanisms of male germ cell differentiation,” BioEssays, vol. 20, no. 7, pp. 555–561, 1998.
[20]  R. Balhorn, M. Cosman, K. Thornton, et al., “Protamine mediated condensation of DNA in mammalian sperm,” in The Male Gamete: From Basic Science to Clinical Applications, C. Gagnon, Ed., pp. 55–70, River Press, 1999.
[21]  R. E. Braun, J. J. Peschon, R. R. Behringer, R. L. Brinster, and R. D. Palmiter, “Protamine 3′-untranslated sequences regulate temporal translational control and subcellular localization of growth hormone in spermatids of transgenic mice,” Genes & Development, vol. 3, no. 6, pp. 793–802, 1989.
[22]  T. A. Brevini-Gandolfi, L. A. Favetta, L. Mauri, A. M. Luciano, F. Cillo, and F. Gandolfi, “Changes in poly(A) tail length of maternal transcripts during in vitro maturation of bovine oocytes and their relation with developmental competence,” Molecular Reproduction and Development, vol. 52, no. 4, pp. 427–433, 1999.
[23]  B. Gold, H. Fujimoto, J. M. Kramer, R. P. Erickson, and N. B. Hecht, “Haploid accumulation and translational control of phosphoglycerate kinase-2 messenger RNA during mouse spermatogenesis,” Developmental Biology, vol. 98, no. 2, pp. 392–399, 1983.
[24]  W. Gu and Y. K. Kwon, “In postmeiotic male germ cells poly (A) shortening accompanies translation of mRNA encoding gamma enteric actin but not cytoplasmic beta and gamma actin mRNAs,” Molecular Reproduction and Development, vol. 44, no. 2, pp. 141–145, 1996.
[25]  H. White-Cooper and I. Davidson, “Unique aspects of transcription regulation in male germ cells,” Cold Spring Harbor Perspectives in Biology, vol. 3, no. 7, 2011.
[26]  B. E. Fischer, E. Wasbrough, L. A. Meadows et al., “Conserved properties of Drosophila and human spermatozoal mRNA repertoires,” Proceedings of the Royal Society B: Biological Sciences, vol. 279, no. 1738, pp. 2636–2644, 2012.
[27]  E. Sendler, G. D. Johnson, and S. A. Krawetz, “Local and global factors affecting RNA sequencing analysis,” Analytical Biochemistry, vol. 419, no. 2, pp. 317–322, 2011.
[28]  G. D. Johnson, E. Sendler, C. Lalancette, R. Hauser, M. P. Diamond, and S. A. Krawetz, “Cleavage of rRNA ensures translational cessation in sperm at fertilization,” Molecular Human Reproduction, vol. 17, no. 12, pp. 721–726, 2011.
[29]  E. Sendler, G. D. Johnson, S. Mao et al., “Stability, delivery and functions of human sperm RNAs at fertilization,” Nucleic Acids Research, vol. 41, no. 7, pp. 4104–4117, 2013.
[30]  B. Sotolongo, T. T. F. Huang, E. Isenberger, and W. S. Ward, “An endogenous nuclease in hamster, mouse, and human spermatozoa cleaves DNA into loop-sized fragments,” Journal of Andrology, vol. 26, no. 2, pp. 272–280, 2005.
[31]  B. Sotolongo, E. Lino, and W. S. Ward, “Ability of hamster spermatozoa to digest their own DNA,” Biology of Reproduction, vol. 69, no. 6, pp. 2029–2035, 2003.
[32]  P. Fang, P. Zeng, Z. Wang, et al., “Estimated diversity of messenger RNAs in each murine spermatozoa and their potential function during early zygotic development,” Biology of Reproduction, vol. 90, no. 5, article 94, 2014.
[33]  J. Kiani and M. Rassoulzadegan, “A load of small RNAs in the sperm-how many bits of hereditary information?” Cell Research, vol. 23, no. 1, pp. 18–19, 2013.
[34]  W.-M. Liu, R. T. K. Pang, P. C. N. Chiu et al., “Sperm-borne microRNA-34c is required for the first cleavage division in mouse,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 2, pp. 490–494, 2012.
[35]  A. Govindaraju, A. Uzun, L. Robertson et al., “Dynamics of microRNAs in bull spermatozoa,” Reproductive Biology and Endocrinology, vol. 10, article 82, 2012.
[36]  S. A. Krawetz, A. Kruger, C. Lalancette et al., “A survey of small RNAs in human sperm,” Human Reproduction, vol. 26, no. 12, pp. 3401–3412, 2011.
[37]  P. C. Au, S. Frankenberg, L. Selwood, and M. Familari, “A novel marsupial pri-miRNA transcript has a putative role in gamete maintenance and defines a vertebrate miRNA cluster paralogous to the miR-15a/miR-16-1 cluster,” Reproduction, vol. 142, no. 4, pp. 539–550, 2011.
[38]  G. Le Trionnaire and D. Twell, “Small RNAs in angiosperm gametophytes: from epigenetics to gamete development,” Genes and Development, vol. 24, no. 11, pp. 1081–1085, 2010.
[39]  F. Bouhallier, N. Allioli, F. Lavial et al., “Role of miR-34c microRNA in the late steps of spermatogenesis,” RNA, vol. 16, no. 4, pp. 720–731, 2010.
[40]  Z. He, M. Kokkinaki, D. Pant, G. I. Gallicano, and M. Dym, “Small RNA molecules in the regulation of spermatogenesis,” Reproduction, vol. 137, no. 6, pp. 901–911, 2009.
[41]  R. Grant-Downton, G. le Trionnaire, R. Schmid et al., “MicroRNA and tasiRNA diversity in mature pollen of Arabidopsis thaliana,” BMC Genomics, vol. 10, article no. 643, 2009.
[42]  R. Grant-Downton, S. Hafidh, D. Twell, and H. G. Dickinson, “Small RNA pathways are present and functional in the angiosperm male gametophyte,” Molecular Plant, vol. 2, no. 3, pp. 500–512, 2009.
[43]  C. Chambers and B. Shuai, “Profiling microRNA expression in Arabidopsis pollen using microRNA array and real-time PCR,” BMC Plant Biology, vol. 9, article 87, 2009.
[44]  E. Marcon, T. Babak, G. Chua, T. Hughes, and P. B. Moens, “miRNA and piRNA localization in the male mammalian meiotic nucleus,” Chromosome Research, vol. 16, no. 2, pp. 243–260, 2008.
[45]  S. T. Grivna, E. Beyret, Z. Wang, and H. Lin, “A novel class of small RNAs in mouse spermatogenic cells,” Genes and Development, vol. 20, no. 13, pp. 1709–1714, 2006.
[46]  G. C. Ostermeier, R. J. Goodrich, J. S. Moldenhauer, M. P. Diamond, and S. A. Krawetz, “A suite of novel human spermatozoal RNAs,” Journal of Andrology, vol. 26, no. 1, pp. 70–74, 2005.
[47]  Y. Gur and H. Breitbart, “Mammalian sperm translate nuclear-encoded proteins by mitochondrial-type ribosomes,” Genes & Development, vol. 20, no. 4, pp. 411–416, 2006.
[48]  C. Zhao, X.-J. Guo, Z.-H. Shi et al., “Role of translation by mitochondrial-type ribosomes during sperm capacitation: an analysis based on a proteomic approach,” Proteomics, vol. 9, no. 5, pp. 1385–1399, 2009.
[49]  D. Ickowicz, M. Finkelstein, and H. Breitbart, “Mechanism of sperm capacitation and the acrosome reaction: role of protein kinases,” Asian Journal of Andrology, vol. 14, no. 6, pp. 816–821, 2012.
[50]  P. E. Visconti, D. Krapf, J. L. de la Vega-Beltrán, J. J. Acevedo, and A. Darszon, “Ion channels, phosphorylation and mammalian sperm capacitation,” Asian Journal of Andrology, vol. 13, no. 3, pp. 395–405, 2011.
[51]  M. A. Baker, “The 'omics revolution and our understanding of sperm cell biology,” Asian Journal of Andrology, vol. 13, no. 1, pp. 6–10, 2011.
[52]  R. R. Zhou, B. Wang, J. Wang, H. Schatten, and Y. Z. Zhang, “Is the mitochondrial cloud the selection machinery for preferentially transmitting wild-type mtDNA between generations? Rewinding müller’s ratchet efficiently,” Current Genetics, vol. 56, no. 2, pp. 101–107, 2010.
[53]  M. Markewitz, S. Graff, and R. J. Veenema, “Absence of RNA synthesis in shed human spermatozoa,” Nature, vol. 214, no. 5086, pp. 402–403, 1967.
[54]  C. Díez-Sánchez, E. Ruiz-Pesini, J. Montoya, A. Pérez-Martos, J. A. Enríquez, and M. J. López-Pérez, “Mitochondria from ejaculated human spermatozoa do not synthesize proteins,” FEBS Letters, vol. 553, no. 1-2, pp. 205–208, 2003.
[55]  D. Miller, P. Z. Tang, C. Skinner, and R. Lilford, “Differential RNA fingerprinting as a tool in the analysis of spermatozoal gene expression,” Human Reproduction, vol. 9, no. 5, pp. 864–869, 1994.
[56]  D. Miller, D. Briggs, H. Snowden et al., “A complex population of RNAs exists in human ejaculate spermatozoa: implications for understanding molecular aspects of spermiogenesis,” Gene, vol. 237, no. 2, pp. 385–392, 1999.
[57]  S. M. Wykes, D. W. Visscher, and S. A. Krawetz, “Haploid transcripts persist in mature human spermatozoa,” Molecular Human Reproduction, vol. 3, no. 1, pp. 15–19, 1997.
[58]  I. I. Concha, U. Urzua, A. Yanez, R. Schroeder, C. Pessot, and L. O. Burzio, “U1 and U2 snRNA are localized in the sperm nucleus,” Experimental Cell Research, vol. 204, no. 2, pp. 378–381, 1993.
[59]  A. H. Balen and H. S. Jacobs, Infertility in Practice, Churchill Livingstone, 2003.
[60]  R. H. Foote, “Value of testicular and sperm profiles in optimizing reproductive success: lessons learned from selective breeding programs of domestic and laboratory animals,” Progress in Clinical and Biological Research, vol. 302, pp. 107–126, 1989.
[61]  K. A. Abraham and P. M. Bhargava, “Nucleic acid metabolism of mammalian spermatozoa,” The Biochemical journal, vol. 86, pp. 298–307, 1963.
[62]  P. M. Bhargava, “Incorporation of radioactive amino-acids in the proteins of bull spermatozoa,” Nature, vol. 179, no. 4570, pp. 1120–1121, 1957.
[63]  F. Martin and J. Brachet, “Autoradiographic studies on the incorporation of amino acids into spermatozoa,” Experimental Cell Research, vol. 17, no. 3, pp. 399–404, 1959.
[64]  J. C. White, I. Leslie, and J. N. Davidson, “Nucleic acids of bone marrow cells, with special reference to pernicious anaemia,” The Journal of Pathology and Bacteriology, vol. 66, no. 1, pp. 291–306, 1953.
[65]  C. M. Mauritzen, A. B. Roy, and E. Stedman, “The ribosenucleic acid content of isolated cell nuclei,” Proceedings of the Royal Society B: Biological Sciences, vol. 140, no. 898, pp. 18–31, 1952.
[66]  J. MacLaughlin and C. Terner, “Ribonucleic acid synthesis by spermatozoa from the rat and hamster,” Biochemical Journal, vol. 133, no. 4, pp. 635–639, 1973.
[67]  E. Premkumar and P. M. Bhargava, “Transcription and translation in bovine spermatozoa,” Nature: New biology, vol. 240, no. 100, pp. 139–143, 1972.
[68]  C. J. Betlach and R. P. Erickson, “28 s and 18 s ribonucleic acid from mammalian spermatozoa.,” Journal of Experimental Zoology, vol. 198, no. 1, pp. 49–55, 1976.
[69]  N. Bissonnette, J.-P. Lévesque-Sergerie, C. Thibault, and G. Boissonneault, “Spermatozoal transcriptome profiling for bull sperm motility: a potential tool to evaluate semen quality,” Reproduction, vol. 138, no. 1, pp. 65–80, 2009.
[70]  S. A. Krawetz, “Paternal contribution: new insights and future challenges,” Nature Reviews Genetics, vol. 6, no. 8, pp. 633–642, 2005.
[71]  H. Cappallo-Obermann, W. Schulze, H. Jastrow, V. Baukloh, and A.-N. Spiess, “Highly purified spermatozoal RNA obtained by a novel method indicates an unusual 28S/18S rRNA ratio and suggests impaired ribosome assembly,” Molecular Human Reproduction, vol. 17, no. 11, pp. 669–678, 2011.
[72]  T. G. Cooper, “Cytoplasmic droplets: the good, the bad or just confusing?” Human Reproduction, vol. 20, no. 1, pp. 9–11, 2005.
[73]  T. G. Cooper and C.-H. Yeung, “Acquisition of volume regulatory response of sperm upon maturation in the epididymis and the role of the cytoplasmic droplet,” Microscopy Research and Technique, vol. 61, no. 1, pp. 28–38, 2003.
[74]  C. J. Card, E. J. Anderson, S. Zamberlan, K. E. Krieger, M. Kaproth, and B. L. Sartini, “Cryopreserved bovine spermatozoal transcript profile as revealed by high-throughput ribonucleic acid sequencing,” Biology of Reproduction, vol. 88, no. 2, article 49, 2013.
[75]  J. M. Feugang, N. Rodriguez-Osorio, A. Kaya et al., “Transcriptome analysis of bull spermatozoa: implications for male fertility,” Reproductive BioMedicine Online, vol. 21, no. 3, pp. 312–324, 2010.
[76]  I. Gilbert, N. Bissonnette, G. Boissonneault, M. Vallée, and C. Robert, “A molecular analysis of the population of mRNA in bovine spermatozoa,” Reproduction, vol. 133, no. 6, pp. 1073–1086, 2007.
[77]  C. Lalancette, C. Thibault, I. Bachand, N. Caron, and N. Bissonnette, “Transcriptome analysis of bull semen with extreme nonreturn rate: Use of suppression-subtractive hybridization to identify functional markers for fertility,” Biology of Reproduction, vol. 78, no. 4, pp. 618–635, 2008.
[78]  Y. Zhao, Q. Li, C. Yao et al., “Characterization and quantification of mRNA transcripts in ejaculated spermatozoa of fertile men by serial analysis of gene expression,” Human Reproduction, vol. 21, no. 6, pp. 1583–1590, 2006.
[79]  C. C. Yang, Y. S. Lin, C. C. Hsu, S. C. Wu, E. C. Lin, and W. T. K. Cheng, “Identification and sequencing of remnant messenger RNAs found in domestic swine (Sus scrofa) fresh ejaculated spermatozoa,” Animal Reproduction Science, vol. 113, no. 1–4, pp. 143–155, 2009.
[80]  P. J. Das, F. McCarthy, M. Vishnoi et al., “Stallion sperm transcriptome comprises functionally coherent coding and regulatory RNAs as revealed by microarray analysis and RNA-seq,” PLoS ONE, vol. 8, no. 2, Article ID e56535, 2013.
[81]  G. C. Ostermeier, D. Miller, J. D. Huntriss, M. P. Diamond, and S. A. Krawetz, “Reproductive biology: delivering spermatozoan RNA to the oocyte,” Nature, vol. 429, no. 6988, article 154, 2004.
[82]  D. Miller, “Analysis and significance of messenger RNA in human ejaculated spermatozoa,” Molecular Reproduction and Development, vol. 56, supplement 2, pp. 259–264, 2000.
[83]  D. Bourc’his and O. Voinnet, “A small-RNA perspective on gametogenesis, fertilization, and early zygotic development,” Science, vol. 330, no. 6004, pp. 617–622, 2010.
[84]  A. E. Peaston, B. B. Knowles, and K. W. Hutchison, “Genome plasticity in the mouse oocyte and early embryo,” Biochemical Society Transactions, vol. 35, part 3, pp. 618–622, 2007.
[85]  T. Kono, Y. Obata, Q. Wu et al., “Birth of parthenogenetic mice that can develop to adulthood,” Nature, vol. 428, no. 6985, pp. 860–864, 2004.
[86]  D. Miller, M. Brinkworth, and D. Iles, “Paternal DNA packaging in spermatozoa: more than the sum of its parts? DNA, histones, protamines and epigenetics,” Reproduction, vol. 139, no. 2, pp. 287–301, 2010.
[87]  M. Pavone, J. Innes, J. Hirshfeld-Cytron, R. Kazer, and J. Zhang, “Comparing thaw survival, implantation and live birth rates from cryopreserved zygotes, embryos and blastocysts,” Journal of Human Reproductive Sciences, vol. 4, no. 1, pp. 23–28, 2011.
[88]  S. Lambard, I. Galeraud-Denis, G. Martin, R. Levy, A. Chocat, and S. Carreau, “Analysis and significance of mRNA in human ejaculated sperm from normozoospermic donors: relationship to sperm motility and capacitation,” Molecular Human Reproduction, vol. 10, no. 7, pp. 535–541, 2004.
[89]  R. Oliva, “Protamines and male infertility,” Human Reproduction Update, vol. 12, no. 4, pp. 417–435, 2006.
[90]  C. Cho, H. Jung-Ha, W. D. Willis et al., “Protamine 2 deficiency leads to sperm DNA damage and embryo death in mice,” Biology of Reproduction, vol. 69, no. 1, pp. 211–217, 2003.
[91]  S. García-Herrero, N. Garrido, J. A. Martínez-Conejero, J. Remohí, A. Pellicer, and M. Meseguer, “Differential transcriptomic profile in spermatozoa achieving pregnancy or not via ICSI,” Reproductive BioMedicine Online, vol. 22, no. 1, pp. 25–36, 2011.
[92]  N. Garrido, J. A. Martínez-Conejero, J. Jauregui et al., “Microarray analysis in sperm from fertile and infertile men without basic sperm analysis abnormalities reveals a significantly different transcriptome,” Fertility and Sterility, vol. 91, no. 4, pp. 1307–1310, 2009.
[93]  WHO, WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction, World Health Organisation, 1999.
[94]  S. Bonache, A. Mata, M. D. Ramos, L. Bassas, and S. Larriba, “Sperm gene expression profile is related to pregnancy rate after insemination and is predictive of low fecundity in normozoospermic men,” Human Reproduction, vol. 27, no. 6, pp. 1556–1567, 2012.
[95]  A. E. Platts, D. J. Dix, H. E. Chemes et al., “Success and failure in human spermatogenesis as revealed by teratozoospermic RNAs,” Human Molecular Genetics, vol. 16, no. 7, pp. 763–773, 2007.
[96]  A. N. Yatsenko, A. Roy, R. Chen et al., “Non-invasive genetic diagnosis of male infertility using spermatozoal RNA: KLHL 10 mutations in oligozoospermic patients impair homodimerization,” Human Molecular Genetics, vol. 15, no. 23, pp. 3411–3419, 2006.
[97]  V. Grandjean, P. Gounon, N. Wagner et al., “The miR-124-Sox9 paramutation: RNA-mediated epigenetic control of embryonic and adult growth,” Development, vol. 136, no. 21, pp. 3647–3655, 2009.
[98]  X. Liang, D. Zhou, C. Wei et al., “MicroRNA-34c enhances murine male germ cell apoptosis through targeting ATF1,” PLoS ONE, vol. 7, no. 3, Article ID e33861, 2012.
[99]  C. P. Concepcion, Y.-C. Han, P. Mu et al., “Intact p53-dependent responses in miR-34-deficient mice,” PLoS Genetics, vol. 8, no. 7, Article ID e1002797, 2012.
[100]  M. D. Anway, A. S. Cupp, N. Uzumcu, and M. K. Skinner, “Epigenetic transgenerational actions of endocrine disruptors and male fertility,” Science, vol. 308, no. 5727, pp. 1466–1469, 2005.
[101]  J. Bender, “RNA-directed DNA methylation: getting a grip on mechanism,” Current Biology, vol. 22, no. 10, pp. R400–R401, 2012.
[102]  C. Belleannée, é. Calvo, J. Caballero, and R. Sullivan, “Epididymosomes convey different repertoires of micrornas throughout the bovine epididymis,” Biology of Reproduction, vol. 89, no. 2, article 30, 2013.
[103]  J. Caballero, G. Frenette, and R. Sullivan, “Post testicular sperm maturational changes in the bull: important role of the epididymosomes and prostasomes,” Veterinary Medicine International, vol. 2011, Article ID 757194, 13 pages, 2011.
[104]  A. Etheridge, C. P. C. Gomes, R. W. Pereira, D. Galas, and K. Wang, “The complexity, function, and applications of RNA in circulation,” Frontiers in Genetics, vol. 4, article 115, 2013.
[105]  A. Dean and R. M. Sharpe, “Anogenital distance or digit length ratio as measures of fetal androgen exposure: relationship to male reproductive development and its disorders,” Journal of Clinical Endocrinology and Metabolism, vol. 98, no. 6, pp. 2230–2238, 2013.
[106]  N. E. Skakkeb?k, E. Rajpert-De Meyts, and K. M. Main, “Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects,” Human Reproduction, vol. 16, no. 5, pp. 972–978, 2001.
[107]  P. M. Foster, E. Mylchreest, K. W. Gaido, and M. Sar, “Effects of phthalate esters on the developing reproductive tract of male rats,” Human Reproduction Update, vol. 7, no. 3, pp. 231–235, 2001.
[108]  J. R. Seckl and M. C. Holmes, “Mechanisms of disease: glucocorticoids, their placental metabolism and fetal 'programming' of adult pathophysiology,” Nature Clinical Practice Endocrinology and Metabolism, vol. 3, no. 6, pp. 479–488, 2007.
[109]  J. Lamarck, Philosophie zoologique ou exposition des considérations relatives à l'histoire naturelle des animaux, University of California Press, 1809.
[110]  R. W. Burkhardt Jr., “Lamarck, evolution, and the inheritance of acquired characters,” Genetics, vol. 194, no. 4, pp. 793–805, 2013.
[111]  W. Reik, K. Davies, W. Dean, G. Kelsey, and M. Constancia, “Imprinted genes and the coordination of fetal and postnatal growth in mammals,” Novartis Foundation Symposium, vol. 237, pp. 19–35, 2001.
[112]  P. Hajkova, S. Erhardt, N. Lane et al., “Epigenetic reprogramming in mouse primordial germ cells,” Mechanisms of Development, vol. 117, no. 1-2, pp. 15–23, 2002.
[113]  C. Pittoggi, L. Renzi, G. Zaccagnini et al., “A fraction of mouse sperm chromatin is organized in nucleosomal hypersensitive domains enriched in retroposon DNA,” Journal of Cell Science, vol. 112, no. 20, pp. 3537–3548, 1999.
[114]  M. Benchaib, V. Braun, D. Ressnikof et al., “Influence of global sperm DNA methylation on IVF results,” Human Reproduction, vol. 20, no. 3, pp. 768–773, 2005.
[115]  S. Houshdaran, V. K. Cortessis, K. Siegmund, A. Yang, P. W. Laird, and R. Z. Sokol, “Widespread epigenetic abnormalities suggest a broad DNA methylation erasure defect in abnormal human sperm,” PLoS ONE, vol. 2, no. 12, Article ID e1289, 2007.
[116]  P. N. Moreira, R. Fernández-González, D. Rizos, M. Ramirez, M. Perez-Crespo, and A. Gutiérrez-Adán, “Inadvertent transgenesis by conventional ICSI in mice,” Human Reproduction, vol. 20, no. 12, pp. 3313–3317, 2005.
[117]  K. Smith and C. Spadafora, “Sperm-mediated gene transfer: applications and implications,” BioEssays, vol. 27, no. 5, pp. 551–562, 2005.
[118]  A. Vargiolu, S. Manzini, M. De Cecco et al., “In vitro production of multigene transgenic blastocysts via sperm-mediated gene transfer allows rapid screening of constructs to be used in xenotransplantation experiments,” Transplantation Proceedings, vol. 42, no. 6, pp. 2142–2145, 2010.
[119]  C. Cossetti, L. Lugini, L. Astrologo, I. Saggio, S. Fais, and C. Spadafora, “Soma-to-germline transmission of RNA in mice xenografted with human tumour cells: possible transport by exosomes,” PloS one, vol. 9, no. 7, Article ID e101629, 2014.
[120]  K. Gapp, L. von Ziegler, R. Y. Tweedie-Cullen, and I. M. Mansuy, “Early life epigenetic programming and transmission of stress-induced traits in mammals: how and when can environmental factors influence traits and their transgenerational inheritance?” BioEssays, vol. 36, no. 5, pp. 491–502, 2014.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413