Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Epigenetic inheritance of acquired traits through sperm RNAs and sperm RNA modifications

Key Points

  • Recent evidence increasingly supports the idea that certain ancestral life experiences acquired in the environment can be inherited by offspring; paternally acquired characteristics can be encoded in the sperm in the form of epigenetic information in addition to DNA sequences.

  • Sperm RNAs, in particular sperm microRNAs (miRNAs) and tRNA-derived small RNAs (tsRNAs), can mediate intergenerational transmission of paternally acquired phenotypes such as diet-induced metabolic disorders and mental stress phenotypes.

  • The mechanisms by which sperm RNAs respond to environmental changes and encode the acquired traits remain unclear but may involve environmental–somatic–germline interactions that may be mediated by extracellular vesicles (EVs) and mobile RNAs, and involve a breach of the somatic–germline barrier.

  • Sperm RNAs may initiate a transcriptional cascade of effects throughout embryonic development to induce a paternally acquired phenotype in offspring; how the initial effects caused by sperm RNAs are converted to a stable form of information to allow transgenerational inheritance remains a major puzzle but possibly involves interplay among transposable elements, DNA methylation and chromatin structure.

  • Emerging evidence suggests that RNA modifications in sperm RNAs have an essential role in modulating epigenetic memory. Novel methods are required to map the locations of multiple RNA modifications in each RNA species, especially for tsRNAs and miRNAs that can induce offspring phenotypes.

  • It remains unknown how many types of acquired traits can be transmitted to offspring through the germ line and under what circumstances this is likely to occur.

Abstract

Once deemed heretical, emerging evidence now supports the notion that the inheritance of acquired characteristics can occur through ancestral exposures or experiences and that certain paternally acquired traits can be 'memorized' in the sperm as epigenetic information. The search for epigenetic factors in mammalian sperm that transmit acquired phenotypes has recently focused on RNAs and, more recently, RNA modifications. Here, we review insights that have been gained from studying sperm RNAs and RNA modifications, and their roles in influencing offspring phenotypes. We discuss the possible mechanisms by which sperm become acquisitive following environmental–somatic–germline interactions, and how they transmit paternally acquired phenotypes by shaping early embryonic development.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Small non-coding RNAs in mouse male germ cells.
Figure 2: Potential mechanisms involving sperm RNAs in early embryos.
Figure 3: Current knowledge of sperm RNA modifications.

Similar content being viewed by others

References

  1. Landman, O. E. The inheritance of acquired characteristics. Annu. Rev. Genet. 25, 1–20 (1991).

    CAS  PubMed  Google Scholar 

  2. Rechavi, O. et al. Starvation-induced transgenerational inheritance of small RNAs in C. elegans. Cell 158, 277–287 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Ost, A. et al. Paternal diet defines offspring chromatin state and intergenerational obesity. Cell 159, 1352–1364 (2014).

    PubMed  Google Scholar 

  4. Ng, S. F. et al. Chronic high-fat diet in fathers programs β-cell dysfunction in female rat offspring. Nature 467, 963–966 (2010).

    CAS  PubMed  Google Scholar 

  5. Carone, B. R. et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 143, 1084–1096 (2010). References 4 and 5 show that acquired metabolic disorders due to paternal diets could be inherited by the offspring.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Dias, B. G. & Ressler, K. J. Parental olfactory experience influences behavior and neural structure in subsequent generations. Nat. Neurosci. 17, 89–96 (2014).

    CAS  PubMed  Google Scholar 

  7. Anway, M. D., Cupp, A. S., Uzumcu, M. & Skinner, M. K. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466–1469 (2005); correction, 328, 690 (2010).

    CAS  PubMed  Google Scholar 

  8. Rodgers, A. B., Morgan, C. P., Bronson, S. L., Revello, S. & Bale, T. L. Paternal stress exposure alters sperm microRNA content and reprograms offspring HPA stress axis regulation. J. Neurosci. 33, 9003–9012 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Chen, Q. et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science 351, 397–400 (2016). This study demonstrates that zygotic injection of sperm total RNAs or tsRNAs from males fed a HFD can induce metabolic disorders in offspring, and that RNA modifications in sperm tsRNAs are sensitive to a HFD.

    CAS  PubMed  Google Scholar 

  10. Huypens, P. et al. Epigenetic germline inheritance of diet-induced obesity and insulin resistance. Nat. Genet. 48, 497–499 (2016).

    CAS  PubMed  Google Scholar 

  11. Sharma, U. et al. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science 351, 391–396 (2016). This paper provides evidence suggesting that sperm can gain tsRNAs during epididymal transition, which can influence embryonic gene expression.

    CAS  PubMed  Google Scholar 

  12. Lane, M., Robker, R. L. & Robertson, S. A. Parenting from before conception. Science 345, 756–760 (2014).

    CAS  PubMed  Google Scholar 

  13. Feng, S., Jacobsen, S. E. & Reik, W. Epigenetic reprogramming in plant and animal development. Science 330, 622–627 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Wei, Y. et al. Paternally induced transgenerational inheritance of susceptibility to diabetes in mammals. Proc. Natl Acad. Sci. USA 111, 1873–1878 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Daxinger, L. & Whitelaw, E. Understanding transgenerational epigenetic inheritance via the gametes in mammals. Nat. Rev. Genet. 13, 153–162 (2012).

    CAS  PubMed  Google Scholar 

  16. Shea, J. M. et al. Genetic and epigenetic variation, but not diet, shape the sperm methylome. Dev. Cell 35, 750–758 (2015). This study suggests that DNA methylation is not the primary epigenetic carrier for diet-induced sperm changes that are responsible for reprogramming of offspring metabolism.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Radford, E. J. et al. In utero effects. In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science 345, 1255903 (2014). This article shows that in utero nutritional exposure during critical windows of germ-cell development alters the sperm methylome of offspring, but the altered methylation does not persist in the next generation.

    PubMed  PubMed Central  Google Scholar 

  18. Iqbal, K. et al. Deleterious effects of endocrine disruptors are corrected in the mammalian germline by epigenome reprogramming. Genome Biol. 16, 59 (2015).

    PubMed  PubMed Central  Google Scholar 

  19. de Waal, E. et al. Primary epimutations introduced during intracytoplasmic sperm injection (ICSI) are corrected by germline-specific epigenetic reprogramming. Proc. Natl Acad. Sci. USA 109, 4163–4168 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hammoud, S. S. et al. Distinctive chromatin in human sperm packages genes for embryo development. Nature 460, 473–478 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Brykczynska, U. et al. Repressive and active histone methylation mark distinct promoters in human and mouse spermatozoa. Nat. Struct. Mol. Biol. 17, 679–687 (2010).

    CAS  PubMed  Google Scholar 

  22. Ragunathan, K., Jih, G. & Moazed, D. Epigenetics. Epigenetic inheritance uncoupled from sequence-specific recruitment. Science 348, 1258699 (2015).

    PubMed  Google Scholar 

  23. Jodar, M. et al. The presence, role and clinical use of spermatozoal RNAs. Hum. Reprod. Update 19, 604–624 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Gapp, K. et al. Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nat. Neurosci. 17, 667–669 (2014). This study provides the first functional evidence for sperm RNAs in transferring the acquired phenotype, showing that the injection of sperm RNAs from a traumatized father induces corresponding phenotypes in offspring.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Rodgers, A. B., Morgan, C. P., Leu, N. A. & Bale, T. L. Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress. Proc. Natl Acad. Sci. USA 112, 13699–13704 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Grandjean, V. et al. RNA-mediated paternal heredity of diet-induced obesity and metabolic disorders. Sci. Rep. 5, 18193 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Kiani, J. et al. RNA-mediated epigenetic heredity requires the cytosine methyltransferase Dnmt2. PLoS Genet. 9, e1003498 (2013). This paper shows that DNMT2 is required for RNA-mediated non-Mendelian epigenetic inheritance in mice.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Nelson, V. R., Heaney, J. D., Tesar, P. J., Davidson, N. O. & Nadeau, J. H. Transgenerational epigenetic effects of the Apobec1 cytidine deaminase deficiency on testicular germ cell tumor susceptibility and embryonic viability. Proc. Natl Acad. Sci. USA 109, E2766–E2773 (2012). This report shows that APOBEC1 is involved in transgenerational epigenetic inheritance of a phenotype.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Bohacek, J. & Mansuy, I. M. Molecular insights into transgenerational non-genetic inheritance of acquired behaviours. Nat. Rev. Genet. 16, 641–652 (2015).

    CAS  PubMed  Google Scholar 

  30. Roemer, I., Reik, W., Dean, W. & Klose, J. Epigenetic inheritance in the mouse. Curr. Biol. 7, 277–280 (1997).

    CAS  PubMed  Google Scholar 

  31. Fullston, T. et al. 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. 27, 4226–4243 (2013).

    CAS  PubMed  Google Scholar 

  32. Wu, L. et al. Paternal psychological stress reprograms hepatic gluconeogenesis in offspring. Cell Metab. 23, 735–743 (2016).

    CAS  PubMed  Google Scholar 

  33. Paris, L. et al. Transgenerational inheritance of enhanced susceptibility to radiation-induced medulloblastoma in newborn Ptch1+/− mice after paternal irradiation. Oncotarget 6, 36098–36112 (2015).

    PubMed  PubMed Central  Google Scholar 

  34. Morgan, H. D., Sutherland, H. G., Martin, D. I. & Whitelaw, E. Epigenetic inheritance at the agouti locus in the mouse. Nat. Genet. 23, 314–318 (1999).

    CAS  PubMed  Google Scholar 

  35. Rakyan, V. K. et al. Transgenerational inheritance of epigenetic states at the murine AxinFu allele occurs after maternal and paternal transmission. Proc. Natl Acad. Sci. USA 100, 2538–2543 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Siklenka, K. et al. Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science 350, aab2006 (2015). This study demonstrates that defects induced by genetically disrupting histone methylation cause transgenerational inheritance of phenotypes in mammals.

    PubMed  Google Scholar 

  37. Ziller, M. J., Hansen, K. D., Meissner, A. & Aryee, M. J. Coverage recommendations for methylation analysis by whole-genome bisulfite sequencing. Nat. Methods 12, 230–232 (2015).

    CAS  PubMed  Google Scholar 

  38. Herman, H. et al. Trans allele methylation and paramutation-like effects in mice. Nat. Genet. 34, 199–202 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Rassoulzadegan, M., Magliano, M. & Cuzin, F. Transvection effects involving DNA methylation during meiosis in the mouse. EMBO J. 21, 440–450 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Hatada, I. et al. Aberrant methylation of an imprinted gene U2af1-rs1(SP2) caused by its own transgene. J. Biol. Chem. 272, 9120–9122 (1997).

    CAS  PubMed  Google Scholar 

  41. Grandjean, V. et al. The miR-124-Sox9 paramutation: RNA-mediated epigenetic control of embryonic and adult growth. Development 136, 3647–3655 (2009).

    CAS  PubMed  Google Scholar 

  42. Wagner, K. D. et al. RNA induction and inheritance of epigenetic cardiac hypertrophy in the mouse. Dev. Cell 14, 962–969 (2008).

    CAS  PubMed  Google Scholar 

  43. Rassoulzadegan, M. et al. RNA-mediated non-Mendelian inheritance of an epigenetic change in the mouse. Nature 441, 469–474 (2006). This article provides the first evidence that sperm RNAs can mediate epigenetic inheritance in mammals.

    CAS  PubMed  Google Scholar 

  44. Chong, S. et al. Modifiers of epigenetic reprogramming show paternal effects in the mouse. Nat. Genet. 39, 614–622 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. de Castro Barbosa, T. et al. High-fat diet reprograms the epigenome of rat spermatozoa and transgenerationally affects metabolism of the offspring. Mol. Metab. 5, 184–197 (2016).

    CAS  PubMed  Google Scholar 

  46. Peng, H. et al. A novel class of tRNA-derived small RNAs extremely enriched in mature mouse sperm. Cell Res. 22, 1609–1612 (2012). This is the first report that tsRNAs are highly enriched in mature sperm.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Schuster, A., Skinner, M. K. & Yan, W. Ancestral vinclozolin exposure alters the epigenetic transgenerational inheritance of sperm small noncoding RNAs. Environ. Epigenet. 2, dvw001 (2016).

    PubMed  PubMed Central  Google Scholar 

  48. Donkin, I. et al. Obesity and bariatric surgery drive epigenetic variation of spermatozoa in humans. Cell Metab. 23, 369–378 (2015).

    PubMed  Google Scholar 

  49. Surani, M. A. Breaking the germ line-soma barrier. Nat. Rev. Mol. Cell Biol. 17, 136 (2016).

    CAS  PubMed  Google Scholar 

  50. Liu, Y. A new perspective on Darwin's pangenesis. Biol. Rev. Camb. Philos. Soc. 83, 141–149 (2008).

    PubMed  Google Scholar 

  51. Koch, S., Acebron, S. P., Herbst, J., Hatiboglu, G. & Niehrs, C. Post-transcriptional Wnt signaling governs epididymal sperm maturation. Cell 163, 1225–1236 (2015).

    CAS  PubMed  Google Scholar 

  52. Sarkies, P. & Miska, E. A. Small RNAs break out: the molecular cell biology of mobile small RNAs. Nat. Rev. Mol. Cell Biol. 15, 525–535 (2014).

    CAS  PubMed  Google Scholar 

  53. Lo Cicero, A., Stahl, P. D. & Raposo, G. Extracellular vesicles shuffling intercellular messages: for good or for bad. Curr. Opin. Cell Biol. 35, 69–77 (2015).

    CAS  PubMed  Google Scholar 

  54. Machtinger, R., Laurent, L. C. & Baccarelli, A. A. Extracellular vesicles: roles in gamete maturation, fertilization and embryo implantation. Hum. Reprod. Update 22, 182–193 (2016).

    CAS  PubMed  Google Scholar 

  55. Mahmoudi, K., Ezrin, A. & Hadjipanayis, C. Small extracellular vesicles as tumor biomarkers for glioblastoma. Mol. Aspects Med. 45, 97–102 (2015).

    CAS  PubMed  Google Scholar 

  56. Vader, P., Mol, E. A., Pasterkamp, G. & Schiffelers, R. M. Extracellular vesicles for drug delivery. Adv. Drug Deliv. Rev. http://dx.doi.org/10.1016/j.addr.2016.02.006 (2016).

  57. Vojtech, L. et al. Exosomes in human semen carry a distinctive repertoire of small non-coding RNAs with potential regulatory functions. Nucleic Acids Res. 42, 7290–7304 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang, Y. et al. Identification and characterization of an ancient class of small RNAs enriched in serum associating with active infection. J. Mol. Cell. Biol. 6, 172–174 (2014).

    PubMed  Google Scholar 

  59. Dhahbi, J. M. et al. 5′ tRNA halves are present as abundant complexes in serum, concentrated in blood cells, and modulated by aging and calorie restriction. BMC Genomics 14, 298 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Li, H., Wu, C., Aramayo, R., Sachs, M. S. & Harlow, M. L. Synaptic vesicles contain small ribonucleic acids (sRNAs) including transfer RNA fragments (trfRNA) and microRNAs (miRNA). Sci. Rep. 5, 14918 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Anderson, P. & Ivanov, P. tRNA fragments in human health and disease. FEBS Lett. 588, 4297–4304 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Cossetti, C. et al. Soma-to-germline transmission of RNA in mice xenografted with human tumour cells: possible transport by exosomes. PLoS ONE 9, e101629 (2014).

    PubMed  PubMed Central  Google Scholar 

  63. Zeybel, M. et al. Multigenerational epigenetic adaptation of the hepatic wound-healing response. Nat. Med. 18, 1369–1377 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Devanapally, S., Ravikumar, S. & Jose, A. M. Double-stranded RNA made in C. elegans neurons can enter the germline and cause transgenerational gene silencing. Proc. Natl Acad. Sci. USA 112, 2133–2138 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Curran, S. P., Wu, X., Riedel, C. G. & Ruvkun, G. A soma-to-germline transformation in long-lived Caenorhabditis elegans mutants. Nature 459, 1079–1084 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Shirayama, M. et al. piRNAs initiate an epigenetic memory of nonself RNA in the C. elegans germline. Cell 150, 65–77 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Buckley, B. A. et al. A nuclear Argonaute promotes multigenerational epigenetic inheritance and germline immortality. Nature 489, 447–451 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Ashe, A. et al. piRNAs can trigger a multigenerational epigenetic memory in the germline of C. elegans. Cell 150, 88–99 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Javurek, A. B. et al. Discovery of a novel seminal fluid microbiome and influence of estrogen receptor alpha genetic status. Sci. Rep. 6, 23027 (2016); corrigendum 6, 25216 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Muraca, M., Putignani, L., Fierabracci, A., Teti, A. & Perilongo, G. Gut microbiota-derived outer membrane vesicles: under-recognized major players in health and disease? Discov. Med. 19, 343–348 (2015).

    PubMed  Google Scholar 

  71. Hoshino, A. et al. Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Ridder, K. et al. Extracellular vesicle-mediated transfer of genetic information between the hematopoietic system and the brain in response to inflammation. PLoS Biol. 12, e1001874 (2014).

    PubMed  PubMed Central  Google Scholar 

  73. Gregor, M. F. & Hotamisligil, G. S. Inflammatory mechanisms in obesity. Annu. Rev. Immunol. 29, 415–445 (2011).

    CAS  PubMed  Google Scholar 

  74. Cohen, S. et al. Chronic stress, glucocorticoid receptor resistance, inflammation, and disease risk. Proc. Natl Acad. Sci. USA 109, 5995–5999 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Fan, Y. et al. Diet-induced obesity in male C57BL/6 mice decreases fertility as a consequence of disrupted blood-testis barrier. PLoS ONE 10, e0120775 (2015).

    PubMed  PubMed Central  Google Scholar 

  76. Nargund, V. H. Effects of psychological stress on male fertility. Nat. Rev. Urol. 12, 373–382 (2015).

    CAS  PubMed  Google Scholar 

  77. Crean, A. J., Kopps, A. M. & Bonduriansky, R. Revisiting telegony: offspring inherit an acquired characteristic of their mother's previous mate. Ecol. Lett. 17, 1545–1552 (2014).

    PubMed  PubMed Central  Google Scholar 

  78. Crean, A. J., Adler, M. I. & Bonduriansky, R. Seminal fluid and mate choice: new predictions. Trends Ecol. Evol. 31, 253–255 (2016).

    PubMed  Google Scholar 

  79. Arroyo, J. D. et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl Acad. Sci. USA 108, 5003–5008 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Feinberg, E. H. & Hunter, C. P. Transport of dsRNA into cells by the transmembrane protein SID-1. Science 301, 1545–1547 (2003).

    CAS  PubMed  Google Scholar 

  81. McEwan, D. L., Weisman, A. S. & Hunter, C. P. Uptake of extracellular double-stranded RNA by SID-2. Mol. Cell 47, 746–754 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Ivanov, P. et al. G-quadruplex structures contribute to the neuroprotective effects of angiogenin-induced tRNA fragments. Proc. Natl Acad. Sci. USA 111, 18201–18206 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Chin, A. R. et al. Cross-kingdom inhibition of breast cancer growth by plant miR159. Cell Res. 26, 217–228 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Yang, J., Farmer, L. M., Agyekum, A. A. & Hirschi, K. D. Detection of dietary plant-based small RNAs in animals. Cell Res. 25, 517–520 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Mlotshwa, S. et al. A novel chemopreventive strategy based on therapeutic microRNAs produced in plants. Cell Res. 25, 521–524 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Zhang, L. et al. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: evidence of cross-kingdom regulation by microRNA. Cell Res. 22, 107–126 (2012).

    CAS  PubMed  Google Scholar 

  87. Zhou, Z. et al. Honeysuckle-encoded atypical microRNA2911 directly targets influenza A viruses. Cell Res. 25, 39–49 (2015).

    CAS  PubMed  Google Scholar 

  88. Ji, L. & Chen, X. Regulation of small RNA stability: methylation and beyond. Cell Res. 22, 624–636 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Skinner, M. K., Guerrero-Bosagna, C. & Haque, M. M. Environmentally induced epigenetic transgenerational inheritance of sperm epimutations promote genetic mutations. Epigenetics 10, 762–771 (2015).

    PubMed  PubMed Central  Google Scholar 

  90. Houri-Ze'evi, L. et al. A tunable mechanism determines the duration of the transgenerational small RNA inheritance in C. elegans. Cell 165, 88–99 (2016).

    CAS  PubMed  Google Scholar 

  91. Piotrowska, K. & Zernicka-Goetz, M. Role for sperm in spatial patterning of the early mouse embryo. Nature 409, 517–521 (2001).

    CAS  PubMed  Google Scholar 

  92. Shi, J. et al. Dynamic transcriptional symmetry-breaking in pre-implantation mammalian embryo development revealed by single-cell RNA-seq. Development 142, 3468–3477 (2015).

    CAS  PubMed  Google Scholar 

  93. Goolam, M. et al. Heterogeneity in Oct4 and Sox2 targets biases cell fate in 4-cell mouse embryos. Cell 165, 61–74 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. White, M. D. et al. Long-lived binding of Sox2 to DNA predicts cell fate in the four-cell mouse embryo. Cell 165, 75–87 (2016). References 92–94 provide the conceptual framework and experimental evidence that the slightest blastomere–blastomere biases during early embryo development can change the trajectory of developmental potential.

    CAS  PubMed  Google Scholar 

  95. Torres-Padilla, M. E., Parfitt, D. E., Kouzarides, T. & Zernicka-Goetz, M. Histone arginine methylation regulates pluripotency in the early mouse embryo. Nature 445, 214–218 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Plachta, N., Bollenbach, T., Pease, S., Fraser, S. E. & Pantazis, P. Oct4 kinetics predict cell lineage patterning in the early mammalian embryo. Nat. Cell Biol. 13, 117–123 (2011).

    CAS  PubMed  Google Scholar 

  97. Davidson, E. H. Emerging properties of animal gene regulatory networks. Nature 468, 911–920 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Holoch, D. & Moazed, D. RNA-mediated epigenetic regulation of gene expression. Nat. Rev. Genet. 16, 71–84 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Watanabe, T. et al. Role for piRNAs and noncoding RNA in de novo DNA methylation of the imprinted mouse Rasgrf1 locus. Science 332, 848–852 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Zuo, Z. et al. Genome-wide analysis reveals origin of transfer RNA genes from tRNA halves. Mol. Biol. Evol. 30, 2087–2098 (2013).

    CAS  PubMed  Google Scholar 

  101. Wu, T. P. et al. DNA methylation on N6-adenine in mammalian embryonic stem cells. Nature 532, 329–333 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Greer, E. L. et al. DNA methylation on N6-adenine in C. elegans. Cell 161, 868–878 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Morris, K. V. & Mattick, J. S. The rise of regulatory RNA. Nat. Rev. Genet. 15, 423–437 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Engreitz, J. M. et al. The Xist lncRNA exploits three-dimensional genome architecture to spread across the X chromosome. Science 341, 1237973 (2013).

    PubMed  PubMed Central  Google Scholar 

  105. Hacisuleyman, E. et al. Topological organization of multichromosomal regions by the long intergenic noncoding RNA Firre. Nat. Struct. Mol. Biol. 21, 198–206 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Simon, M. D. et al. High-resolution Xist binding maps reveal two-step spreading during X-chromosome inactivation. Nature 504, 465–469 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Xiang, J. F. et al. Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus. Cell Res. 24, 513–531 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Dekker, J. & Mirny, L. The 3D genome as moderator of chromosomal communication. Cell 164, 1110–1121 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Le Thomas, A. et al. Transgenerationally inherited piRNAs trigger piRNA biogenesis by changing the chromatin of piRNA clusters and inducing precursor processing. Genes Dev. 28, 1667–1680 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Fu, Y., Dominissini, D., Rechavi, G. & He, C. Gene expression regulation mediated through reversible m6A RNA methylation. Nat. Rev. Genet. 15, 293–306 (2014).

    CAS  PubMed  Google Scholar 

  111. Chen, K., Zhao, B. S. & He, C. Nucleic acid modifications in regulation of gene expression. Cell Chem. Biol. 23, 74–85 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Yan, M. et al. A high-throughput quantitative approach reveals more small RNA modifications in mouse liver and their correlation with diabetes. Anal. Chem. 85, 12173–12181 (2013).

    CAS  PubMed  Google Scholar 

  113. Machnicka, M. A. et al. MODOMICS: a database of RNA modification pathways — 2013 update. Nucleic Acids Res. 41, D262–D267 (2013).

    CAS  PubMed  Google Scholar 

  114. Cantara, W. A. et al. The RNA Modification Database, RNAMDB: 2011 update. Nucleic Acids Res. 39, D195–D201 (2011).

    CAS  PubMed  Google Scholar 

  115. Schaefer, M. et al. RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes Dev. 24, 1590–1595 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Goll, M. G. et al. Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2. Science 311, 395–398 (2006).

    CAS  PubMed  Google Scholar 

  117. Tuorto, F. et al. RNA cytosine methylation by Dnmt2 and NSun2 promotes tRNA stability and protein synthesis. Nat. Struct. Mol. Biol. 19, 900–905 (2012).

    CAS  PubMed  Google Scholar 

  118. Popow, J., Jurkin, J., Schleiffer, A. & Martinez, J. Analysis of orthologous groups reveals archease and DDX1 as tRNA splicing factors. Nature 511, 104–107 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Han, C. et al. The RNA-binding protein DDX1 promotes primary microRNA maturation and inhibits ovarian tumor progression. Cell Rep. 8, 1447–1460 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Hildebrandt, M. R., Germain, D. R., Monckton, E. A., Brun, M. & Godbout, R. Ddx1 knockout results in transgenerational wild-type lethality in mice. Sci. Rep. 5, 9829 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Zheng, G. et al. Efficient and quantitative high-throughput tRNA sequencing. Nat. Methods 12, 835–837 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Cozen, A. E. et al. ARM-seq: AlkB-facilitated RNA methylation sequencing reveals a complex landscape of modified tRNA fragments. Nat. Methods 12, 879–884 (2015). References 121 and 122 report novel methods to discover previously undetected tRNAs and tsRNAs by RNA-seq.

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Kirchner, S. & Ignatova, Z. Emerging roles of tRNA in adaptive translation, signalling dynamics and disease. Nat. Rev. Genet. 16, 98–112 (2015).

    CAS  PubMed  Google Scholar 

  124. Schwartzman, O. & Tanay, A. Single-cell epigenomics: techniques and emerging applications. Nat. Rev. Genet. 16, 716–726 (2015).

    CAS  PubMed  Google Scholar 

  125. Angermueller, C. et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat. Methods 13, 229–232 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Hou, Y. et al. Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res. 26, 304–319 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Ostermeier, G. C., Dix, D. J., Miller, D., Khatri, P. & Krawetz, S. A. Spermatozoal RNA profiles of normal fertile men. Lancet 360, 772–777 (2002).

    CAS  PubMed  Google Scholar 

  128. Ostermeier, G. C., Miller, D., Huntriss, J. D., Diamond, M. P. & Krawetz, S. A. Reproductive biology: delivering spermatozoan RNA to the oocyte. Nature 429, 154 (2004).

    CAS  PubMed  Google Scholar 

  129. Yuan, S. et al. mir-34b/c and mir-449a/b/c are required for spermatogenesis, but not for the first cleavage division in mice. Biol. Open 4, 212–223 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Liu, W. M. et al. Sperm-borne microRNA-34c is required for the first cleavage division in mouse. Proc. Natl Acad. Sci. USA 109, 490–494 (2012).

    CAS  PubMed  Google Scholar 

  131. Yuan, S. et al. Sperm-borne miRNAs and endo-siRNAs are important for fertilization and preimplantation embryonic development. Development 143, 635–647 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Zhong, C. et al. Parthenogenetic haploid embryonic stem cells efficiently support mouse generation by oocyte injection. Cell Res. 26, 131–134 (2016).

    PubMed  Google Scholar 

  133. Li, Z. et al. Birth of fertile bimaternal offspring following intracytoplasmic injection of parthenogenetic haploid embryonic stem cells. Cell Res. 26, 135–138 (2016).

    PubMed  Google Scholar 

  134. Miller, D. Confrontation, consolidation, and recognition: the oocyte's perspective on the incoming sperm. Cold Spring Harb. Perspect. Med. 5, a023408 (2015).

    PubMed  PubMed Central  Google Scholar 

  135. Chandler, V. L. Paramutation: from maize to mice. Cell 128, 641–645 (2007).

    CAS  PubMed  Google Scholar 

  136. Yuan, S., Oliver, D., Schuster, A., Zheng, H. & Yan, W. Breeding scheme and maternal small RNAs affect the efficiency of transgenerational inheritance of a paramutation in mice. Sci. Rep. 5, 9266 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Beale, L. S. Pangenesis. Nature 4, 25–26 (1871).

    Google Scholar 

  138. Eberhard, W. G. Postcopulatory sexual selection: Darwin's omission and its consequences. Proc. Natl Acad. Sci. USA 106 (Suppl. 1), 10025–10032 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Pizzari, T. & Foster, K. R. Sperm sociality: cooperation, altruism, and spite. PLoS Biol. 6, e130 (2008).

    PubMed  PubMed Central  Google Scholar 

  140. Edward, D. A., Stockley, P. & Hosken, D. J. Sexual conflict and sperm competition. Cold Spring Harb. Perspect. Biol. 7, a017707 (2015).

    PubMed Central  Google Scholar 

Download references

Acknowledgements

The Chen laboratory is currently supported by start-up funds from the University of Nevada, Reno School of Medicine, USA. This work is in part funded by the Ministry of Science and Technology of the People's Republic of China (2015CB943000 to Q.C. and 2016YFA0500903, 2015DFG32640 to E.D.), the National Natural Science Foundation of China (81490742, 31671568 to E.D.), the US National Institutes of Health (HD060858, HD071736 and HD085506 to W.Y.) and the John Templeton Foundation, USA (PID: 50183 to W.Y.). The authors thank all Chen laboratory members for critical discussions on the contents of the manuscript and for help in preparing the graphics and illustrations.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Qi Chen, Wei Yan or Enkui Duan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

PowerPoint slides

Glossary

Parthenogenetic

A type of asexual reproduction that occurs when a female gamete develops a new individual without being fertilized by a male gamete

RNA-editing events

Molecular processes by which specific nucleotide sequences in an RNA molecule are changed, such as C-to-U and A-to-I editing.

Kinky-tail phenotype

A mouse phenotype characterized by a kinked tail containing a sharp bend at an angle to the main tail axis.

Transvection

An epigenetic phenomenon that involves the interaction between two homologous chromosomes, resulting in either gene activation or repression at an allele.

Paramutation

Interaction between two alleles at a single locus during meiosis, resulting in epigenetic transfer of information from one allele to another that is heritable for generations.

Epididymal maturation

Spermatozoa from testis undergo a maturation process during transit from the proximal to the distal end of the epididymis, acquiring motility and fertility.

Polyandrous

The mating behaviour of animals in which the females mate with more than one male in a single breeding cycle.

G-Quadruplex conformation

Guanine-rich oligonucleotides that assembled into intra- or inter-molecular guanine tetrad structures, which can be formed by both DNA and RNA.

Lamarckism

The idea that an organism can pass on characteristics that are acquired during its lifetime to its offspring.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Q., Yan, W. & Duan, E. Epigenetic inheritance of acquired traits through sperm RNAs and sperm RNA modifications. Nat Rev Genet 17, 733–743 (2016). https://doi.org/10.1038/nrg.2016.106

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg.2016.106

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing