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:

RNA-mediated epigenetic regulation of gene expression

Key Points

  • Small RNAs, which function within Argonaute (AGO) complexes that target nascent transcript scaffolds, and long non-coding RNAs (lncRNAs), or even mRNAs, that themselves act as chromatin-associated scaffolds recruit chromatin-modifying activities to mediate stable changes in gene expression.

  • Small RNAs maintain a persistent memory of epigenetic silencing by forming positive feedback loops with chromatin-based signals. These self-reinforcing loops are best understood in the pericentromeric heterochromatin of Schizosaccharomyces pombe and the RNA-directed DNA methylation pathway of Arabidopsis thaliana.

  • In Drosophila melanogaster, recent evidence suggests the existence of a self-reinforcing relationship between Piwi-interacting RNA (piRNA) biogenesis and histone H3 lysine 9 (H3K9) methylation at piRNA source loci. In mice, piRNAs direct DNA methylation, but these signals do not seem to form a self-reinforcing loop.

  • In Caenorhabditis elegans, the AGO protein HRDE-1 maintains heritable silencing of foreign sequences after recognition by the piRNA pathway, while the AGO protein CSR-1 protects 'self' sequences from silencing.

  • Numerous lncRNAs have been proposed to directly recruit the Polycomb repressive complex 2 (PRC2) histone methyltransferase, but a lack of specificity in PRC2–RNA interactions suggests a more complicated picture.

  • The well-studied lncRNA X inactive specific transcript X (XIST) spreads along the inactive X chromosome and mediates gene silencing by recruiting PRC2. Recent studies show that this occurs indirectly through the RNA-binding protein JARID2.

  • lncRNAs transcribed from enhancer regions can activate gene expression by acting as scaffolds that bring enhancer and promoter regions into proximity, while also recruiting co-activators to modify histones.

  • The mRNAs of meiotic genes in S. pombe are silenced during vegetative growth in a process that involves (but does not require) recruitment of H3K9 methylation activity by the mRNA transcripts themselves. This mechanism may represent an ancestral step in the evolution of lncRNAs, in which RNAs have acquired a scaffold function to recruit histone modifiers but have not yet lost protein-coding potential.

Abstract

Diverse classes of RNA, ranging from small to long non-coding RNAs, have emerged as key regulators of gene expression, genome stability and defence against foreign genetic elements. Small RNAs modify chromatin structure and silence transcription by guiding Argonaute-containing complexes to complementary nascent RNA scaffolds and then mediating the recruitment of histone and DNA methyltransferases. In addition, recent advances suggest that chromatin-associated long non-coding RNA scaffolds also recruit chromatin-modifying complexes independently of small RNAs. These co-transcriptional silencing mechanisms form powerful RNA surveillance systems that detect and silence inappropriate transcription events, and provide a memory of these events via self-reinforcing epigenetic loops.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: The 'nascent transcript' model and a self-reinforcing epigenetic loop in S. pombe.
Figure 2: A self-reinforcing loop linking siRNAs to DNA and histone methylation in A. thaliana.
Figure 3: Small-RNA-driven transcriptional silencing of gene expression in C. elegans.
Figure 4: RNAs, both short and long, represent an alternative to DNA-binding proteins as specificity determinants for epigenetic regulation of gene expression.

Similar content being viewed by others

References

  1. Moazed, D. Small RNAs in transcriptional gene silencing and genome defence. Nature 457, 413–420 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Cech, T. R. & Steitz, J. A. The noncoding RNA revolution — trashing old rules to forge new ones. Cell 157, 77–94 (2014).

    CAS  PubMed  Google Scholar 

  3. Hammond, S. M., Boettcher, S., Caudy, A. A., Kobayashi, R. & Hannon, G. J. Argonaute2, a link between genetic and biochemical analyses of RNAi. Science 293, 1146–1150 (2001).

    CAS  PubMed  Google Scholar 

  4. Hutvagner, G. & Zamore, P. D. A microRNA in a multiple-turnover RNAi enzyme complex. Science 297, 2056–2060 (2002).

    CAS  PubMed  Google Scholar 

  5. Meister, G. et al. Human Argonaute2 mediates RNA cleavage targeted by miRNAs and siRNAs. Mol. Cell 15, 185–197 (2004).

    CAS  PubMed  Google Scholar 

  6. Hamilton, A. J. & Baulcombe, D. C. A species of small antisense RNA in posttranscriptional gene silencing in plants. Science 286, 950–952 (1999).

    CAS  PubMed  Google Scholar 

  7. Dalmay, T., Hamilton, A., Mueller, E. & Baulcombe, D. C. Potato virus X amplicons in Arabidopsis mediate genetic and epigenetic gene silencing. Plant Cell 12, 369–379 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Jones, L. et al. RNA–DNA interactions and DNA methylation in post-transcriptional gene silencing. Plant Cell 11, 2291–2301 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Mette, M. F., van der Winden, J., Matzke, M. A. & Matzke, A. J. Production of aberrant promoter transcripts contributes to methylation and silencing of unlinked homologous promoters in trans. EMBO J. 18, 241–248 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Volpe, T. A. et al. Regulation of heterochromatic silencing and histone H3 lysine-9 methylation by RNAi. Science 297, 1833–1837 (2002).

    CAS  PubMed  Google Scholar 

  11. Reinhart, B. J. & Bartel, D. P. Small RNAs correspond to centromere heterochromatic repeats. Science 297, 1831 (2002).

    CAS  PubMed  Google Scholar 

  12. Matzke, M., Matzke, A. J. & Kooter, J. M. RNA: guiding gene silencing. Science 293, 1080–1083 (2001).

    CAS  PubMed  Google Scholar 

  13. Fire, A. et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391, 806–811 (1998).

    CAS  PubMed  Google Scholar 

  14. Mochizuki, K., Fine, N. A., Fujisawa, T. & Gorovsky, M. A. Analysis of a PIWI-related gene implicates small RNAs in genome rearrangement in tetrahymena. Cell 110, 689–699 (2002). References 6–8, 10, 11 and 14 provided the first evidence for the role of small RNAs in chromatin modification and transcriptional gene silencing.

    CAS  PubMed  Google Scholar 

  15. Brennecke, J. et al. An epigenetic role for maternally inherited piRNAs in transposon silencing. Science 322, 1387–1392 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Guang, S. et al. Small regulatory RNAs inhibit RNA polymerase II during the elongation phase of transcription. Nature 465, 1097–1101 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Gu, S. G. et al. Amplification of siRNA in Caenorhabditis elegans generates a transgenerational sequence-targeted histone H3 lysine 9 methylation footprint. Nature Genet. 44, 157–164 (2012). This study provides convincing evidence that classical RNAi in C. elegans results in specific H3K9 methylation at targeted loci.

    CAS  PubMed  Google Scholar 

  18. Rinn, J. L. & Chang, H. Y. Genome regulation by long noncoding RNAs. Annu. Rev. Biochem. 81, 145–166 (2012).

    CAS  PubMed  Google Scholar 

  19. Lee, J. T. & Bartolomei, M. S. X-inactivation, imprinting, and long noncoding RNAs in health and disease. Cell 152, 1308–1323 (2013).

    CAS  PubMed  Google Scholar 

  20. Schulz, E. G. & Heard, E. Role and control of X chromosome dosage in mammalian development. Curr. Opin. Genet. Dev. 23, 109–115 (2013).

    CAS  PubMed  Google Scholar 

  21. Yang, P. K. & Kuroda, M. I. Noncoding RNAs and intranuclear positioning in monoallelic gene expression. Cell 128, 777–786 (2007).

    CAS  PubMed  Google Scholar 

  22. Bonasio, R. & Shiekhattar, R. Regulation of transcription by long noncoding RNAs. Annu. Rev. Genet. 48, 433–455 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Verdel, A. et al. RNAi-mediated targeting of heterochromatin by the RITS complex. Science 303, 672–676 (2004). This study identifies a nuclear AGO complex, RITS, which uses Dicer-generated small RNAs to target specific chromosome regions for heterochromatin formation. The RITS complex provided the first direct physical link between RNAi and heterochromatin.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Partridge, J. F., Scott, K. S., Bannister, A. J., Kouzarides, T. & Allshire, R. C. Cis-acting DNA from fission yeast centromeres mediates histone H3 methylation and recruitment of silencing factors and cohesin to an ectopic site. Curr. Biol. 12, 1652–1660 (2002).

    CAS  PubMed  Google Scholar 

  25. El-Shami, M. et al. Reiterated WG–GW motifs form functionally and evolutionarily conserved Argonaute-binding platforms in RNAi-related components. Genes Dev. 21, 2539–2544 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Till, S. et al. A conserved motif in Argonaute-interacting proteins mediates functional interactions through the Argonaute PIWI domain. Nature Struct. Mol. Biol. 14, 897–903 (2007).

    CAS  Google Scholar 

  27. Motamedi, M. R. et al. Two RNAi complexes, RITS and RDRC, physically interact and localize to noncoding centromeric RNAs. Cell 119, 789–802 (2004). This study reports the first usage of RNA immunoprecipitation experiments for a chromatin-associated protein.

    CAS  PubMed  Google Scholar 

  28. Buhler, M., Verdel, A. & Moazed, D. Tethering RITS to a nascent transcript initiates RNAi- and heterochromatin-dependent gene silencing. Cell 125, 873–886 (2006).

    CAS  PubMed  Google Scholar 

  29. Buhler, M., Haas, W., Gygi, S. P. & Moazed, D. RNAi-dependent and -independent RNA turnover mechanisms contribute to heterochromatic gene silencing. Cell 129, 707–721 (2007). This was the first study to demonstrate a major role for co-transcriptional RNAi-, and TRAMP- and exosome-dependent RNA degradation in heterochromatic gene silencing. Together with references 27 and 28, it provides evidence for nascent non-coding RNAs as scaffolds for the assembly of RITS and other complexes that mediate heterochromatin formation.

    CAS  PubMed  Google Scholar 

  30. Motamedi, M. R. et al. HP1 proteins form distinct complexes and mediate heterochromatic gene silencing by nonoverlapping mechanisms. Mol. Cell 32, 778–790 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Sugiyama, T. et al. SHREC, an effector complex for heterochromatic transcriptional silencing. Cell 128, 491–504 (2007).

    CAS  PubMed  Google Scholar 

  32. Moazed, D. Mechanisms for the inheritance of chromatin states. Cell 146, 510–518 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Nakayama, J., Rice, J. C., Strahl, B. D., Allis, C. D. & Grewal, S. I. Role of histone H3 lysine 9 methylation in epigenetic control of heterochromatin assembly. Science 292, 110–113 (2001).

    CAS  PubMed  Google Scholar 

  34. Hong, E. J., Villen, J., Gerace, E. L., Gygi, S. P. & Moazed, D. A cullin E3 ubiquitin ligase complex associates with Rik1 and the Clr4 histone H3-K9 methyltransferase and is required for RNAi-mediated heterochromatin formation. RNA Biol. 2, 106–111 (2005).

    CAS  PubMed  Google Scholar 

  35. Horn, P. J., Bastie, J. N. & Peterson, C. L. A Rik1-associated, cullin-dependent E3 ubiquitin ligase is essential for heterochromatin formation. Genes Dev. 19, 1705–1714 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Jia, S., Kobayashi, R. & Grewal, S. I. Ubiquitin ligase component Cul4 associates with Clr4 histone methyltransferase to assemble heterochromatin. Nature Cell Biol. 7, 1007–1013 (2005).

    CAS  PubMed  Google Scholar 

  37. Li, F. et al. Two novel proteins, dos1 and dos2, interact with rik1 to regulate heterochromatic RNA interference and histone modification. Curr. Biol. 15, 1448–1457 (2005).

    CAS  PubMed  Google Scholar 

  38. Bayne, E. H. et al. Stc1: a critical link between RNAi and chromatin modification required for heterochromatin integrity. Cell 140, 666–677 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Gerace, E. L., Halic, M. & Moazed, D. The methyltransferase activity of Clr4Suv39h triggers RNAi independently of histone H3K9 methylation. Mol. Cell 39, 360–372 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhang, K., Mosch, K., Fischle, W. & Grewal, S. I. Roles of the Clr4 methyltransferase complex in nucleation, spreading and maintenance of heterochromatin. Nature Struct. Mol. Biol. (2008).

  41. Li, F., Martienssen, R. & Cande, W. Z. Coordination of DNA replication and histone modification by the Rik1–Dos2 complex. Nature 475, 244–248 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Zaratiegui, M. et al. RNAi promotes heterochromatic silencing through replication-coupled release of RNA Pol II. Nature 479, 135–138 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bayne, E. H. et al. Splicing factors facilitate RNAi-directed silencing in fission yeast. Science 322, 602–606 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Chinen, M., Morita, M., Fukumura, K. & Tani, T. Involvement of the spliceosomal U4 small nuclear RNA in heterochromatic gene silencing at fission yeast centromeres. J. Biol. Chem. 285, 5630–5638 (2010).

    CAS  PubMed  Google Scholar 

  45. Kallgren, S. P. et al. The proper splicing of RNAi factors is critical for pericentric heterochromatin assembly in fission yeast. PLoS Genet. 10, e1004334 (2014).

    PubMed  PubMed Central  Google Scholar 

  46. Selker, E. U. Trichostatin A causes selective loss of DNA methylation in Neurospora. Proc. Natl Acad. Sci. USA 95, 9430–9435 (1998). This paper was the first to propose self-reinforcing relationships between different types of chromatin modifications: DNA methylation and histone deacetylation.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Selker, E. U. Gene silencing: repeats that count. Cell 97, 157–160 (1999).

    CAS  PubMed  Google Scholar 

  48. Richards, E. J. & Elgin, S. C. Epigenetic codes for heterochromatin formation and silencing: rounding up the usual suspects. Cell 108, 489–500 (2002).

    CAS  PubMed  Google Scholar 

  49. Noma, K. et al. RITS acts in cis to promote RNA interference-mediated transcriptional and post-transcriptional silencing. Nature Genet. 36, 1174–1180 (2004).

    CAS  PubMed  Google Scholar 

  50. Colmenares, S. U., Buker, S. M., Buhler, M., Dlakic, M. & Moazed, D. Coupling of double-stranded RNA synthesis and siRNA generation in fission yeast RNAi. Mol. Cell 27, 449–461 (2007).

    CAS  PubMed  Google Scholar 

  51. Hayashi, A. et al. Heterochromatin protein 1 homologue Swi6 acts in concert with Ers1 to regulate RNAi-directed heterochromatin assembly. Proc. Natl Acad. Sci. USA 109, 6159–6164 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Rougemaille, M. et al. Ers1 links HP1 to RNAi. Proc. Natl Acad. Sci. USA 109, 11258–11263 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Freitag, M. et al. DNA methylation is independent of RNA interference in Neurospora. Science 304, 1939 (2004).

    CAS  PubMed  Google Scholar 

  54. Honda, S. & Selker, E. U. Direct interaction between DNA methyltransferase DIM-2 and HP1 is required for DNA methylation in Neurospora crassa. Mol. Cell. Biol. 28, 6044–6055 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Dang, Y., Li, L., Guo, W., Xue, Z. & Liu, Y. Convergent transcription induces dynamic DNA methylation at disiRNA loci. PLoS Genet. 9, e1003761 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Lee, H. C. et al. The DNA/RNA-dependent RNA polymerase QDE-1 generates aberrant RNA and dsRNA for RNAi in a process requiring replication protein A and a DNA helicase. PLoS Biol. 8, e1000496 (2010).

    PubMed  PubMed Central  Google Scholar 

  57. Ream, T. S. et al. Subunit compositions of the RNA-silencing enzymes Pol IV and Pol V reveal their origins as specialized forms of RNA polymerase II. Mol. Cell 33, 192–203 (2009).

    CAS  PubMed  Google Scholar 

  58. Matzke, M. A. & Mosher, R. A. RNA-directed DNA methylation: an epigenetic pathway of increasing complexity. Nature Rev. Genet. 15, 394–408 (2014).

    CAS  PubMed  Google Scholar 

  59. Haag, J. R. et al. In vitro transcription activities of Pol IV, Pol V, and RDR2 reveal coupling of Pol IV and RDR2 for dsRNA synthesis in plant RNA silencing. Mol. Cell 48, 811–818 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Ye, R. et al. Cytoplasmic assembly and selective nuclear import of Arabidopsis Argonaute4/siRNA complexes. Mol. Cell 46, 859–870 (2012).

    CAS  PubMed  Google Scholar 

  61. Wierzbicki, A. T., Haag, J. R. & Pikaard, C. S. Noncoding transcription by RNA polymerase Pol IVb/Pol V mediates transcriptional silencing of overlapping and adjacent genes. Cell 135, 635–648 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Wierzbicki, A. T., Ream, T. S., Haag, J. R. & Pikaard, C. S. RNA polymerase V transcription guides ARGONAUTE4 to chromatin. Nature Genet. 41, 630–634 (2009). This paper provided the first evidence that nascent Pol V transcripts act as scaffolds for the recruitment of AGO4.

    CAS  PubMed  Google Scholar 

  63. He, X. J. et al. NRPD4, a protein related to the RPB4 subunit of RNA polymerase II, is a component of RNA polymerases IV and V and is required for RNA-directed DNA methylation. Genes Dev. 23, 318–330 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Gao, Z. et al. An RNA polymerase II- and AGO4-associated protein acts in RNA-directed DNA methylation. Nature 465, 106–109 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhong, X. et al. Molecular mechanism of action of plant DRM de novo DNA methyltransferases. Cell 157, 1050–1060 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Blevins, T. et al. A two-step process for epigenetic inheritance in Arabidopsis. Mol. Cell 54, 30–42 (2014). This paper shows that histone deacetylation and DNA methylation mark a locus for epigenetic inheritance and siRNA-mediated silencing.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Johnson, L. M. et al. The SRA methyl-cytosine-binding domain links DNA and histone methylation. Curr. Biol. 17, 379–384 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Law, J. A. et al. Polymerase IV occupancy at RNA-directed DNA methylation sites requires SHH1. Nature 498, 385–389 (2013). This paper shows that SHH1 recognizes H3 tails with unmodified lysine 4 and methylated lysine 9 residues, and recruits Pol IV to chromatin to promote siRNA generation. This provides a clear basis for self-reinforcement between histone modification and small RNA biogenesis in plants.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhang, H. et al. DTF1 is a core component of RNA-directed DNA methylation and may assist in the recruitment of Pol IV. Proc. Natl Acad. Sci. USA 110, 8290–8295 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Johnson, L. M., Law, J. A., Khattar, A., Henderson, I. R. & Jacobsen, S. E. SRA-domain proteins required for DRM2-mediated de novo DNA methylation. PLoS Genet. 4, e1000280 (2008).

    PubMed  PubMed Central  Google Scholar 

  71. Johnson, L. M. et al. SRA- and SET-domain-containing proteins link RNA polymerase V occupancy to DNA methylation. Nature 507, 124–128 (2014). This study shows that the putative methyl DNA-binding proteins SUVH2 and SUVH9 recruit Pol V to sites of pre-existing DNA methylation. By promoting further RNA-directed DNA methylation, SUVH2 and SUVH9 thus act as key members of a self-reinforcing loop.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Liu, Z. W. et al. The SET domain proteins SUVH2 and SUVH9 are required for Pol V occupancy at RNA-directed DNA methylation loci. PLoS Genet. 10, e1003948 (2014).

    PubMed  PubMed Central  Google Scholar 

  73. Malone, C. D. & Hannon, G. J. Small RNAs as guardians of the genome. Cell 136, 656–668 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Batista, P. J. et al. PRG-1 and 21U-RNAs interact to form the piRNA complex required for fertility in C. elegans. Mol. Cell 31, 67–78 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Das, P. P. et al. PIWI and piRNAs act upstream of an endogenous siRNA pathway to suppress Tc3 transposon mobility in the Caenorhabditis elegans germline. Mol. Cell 31, 79–90 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Huang, X. A. et al. A major epigenetic programming mechanism guided by piRNAs. Dev. Cell 24, 502–516 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Le Thomas, A. et al. Piwi induces piRNA-guided transcriptional silencing and establishment of a repressive chromatin state. Genes Dev. 27, 390–399 (2013). Together with reference 76, this paper demonstrates that ectopic targeting of Piwi in D. melanogaster using artificially generated piRNAs leads to H3K9 methylation and transcriptional silencing at target loci. This suggests that piRNA-progammed Piwi can interact with nascent transcripts to induce chromatin changes.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Rozhkov, N. V., Hammell, M. & Hannon, G. J. Multiple roles for Piwi in silencing Drosophila transposons. Genes Dev. 27, 400–412 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Sienski, G., Donertas, D. & Brennecke, J. Transcriptional silencing of transposons by PIWI and maelstrom and its impact on chromatin state and gene expression. Cell 151, 964–980 (2012). This study was the first to show that, in D. melanogaster , Piwi and piRNAs silence transposons at the transcriptional level genome-wide.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Wang, S. H. & Elgin, S. C. Drosophila Piwi functions downstream of piRNA production mediating a chromatin-based transposon silencing mechanism in female germ line. Proc. Natl Acad. Sci. USA 108, 21164–21169 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Brower-Toland, B. et al. Drosophila Piwi associates with chromatin and interacts directly with HP1a. Genes Dev. 21, 2300–2311 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Mohn, F., Sienski, G., Handler, D. & Brennecke, J. The Rhino–Deadlock–Cutoff complex licenses noncanonical transcription of dual-strand piRNA clusters in Drosophila. Cell 157, 1364–1379 (2014).

    CAS  PubMed  Google Scholar 

  83. 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 

  84. Aravin, A. A., Hannon, G. J. & Brennecke, J. The Piwi–piRNA pathway provides an adaptive defense in the transposon arms race. Science 318, 761–764 (2007).

    CAS  PubMed  Google Scholar 

  85. Aravin, A. A. et al. A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice. Mol. Cell 31, 785–799 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Carmell, M. A. et al. MIWI2 is essential for spermatogenesis and repression of transposons in the mouse male germline. Dev. Cell 12, 503–514 (2007).

    CAS  PubMed  Google Scholar 

  87. Kuramochi-Miyagawa, S. et al. DNA methylation of retrotransposon genes is regulated by PIWI family members MILI and MIWI2 in murine fetal testes. Genes Dev. 22, 908–917 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. 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 

  89. De Fazio, S. et al. The endonuclease activity of Mili fuels piRNA amplification that silences LINE1 elements. Nature 480, 259–263 (2011).

    CAS  PubMed  Google Scholar 

  90. de Vanssay, A. et al. Paramutation in Drosophila linked to emergence of a piRNA-producing locus. Nature 490, 112–115 (2012). This study reports the first example of paramutation in animals and describes how a locus can become included in the piRNA repertoire. Together with reference 16, it illustrates how small RNAs can serve as the carriers of epigenetic signals.

    CAS  PubMed  Google Scholar 

  91. Chandler, V. L. Paramutation's properties and puzzles. Science 330, 628–629 (2010).

    CAS  PubMed  Google Scholar 

  92. Yigit, E. et al. Analysis of the C. elegans Argonaute family reveals that distinct Argonautes act sequentially during RNAi. Cell 127, 747–757 (2006).

    CAS  PubMed  Google Scholar 

  93. Guang, S. et al. An Argonaute transports siRNAs from the cytoplasm to the nucleus. Science 321, 537–541 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Burkhart, K. B. et al. A pre-mRNA-associating factor links endogenous siRNAs to chromatin regulation. PLoS Genet. 7, e1002249 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Burton, N. O., Burkhart, K. B. & Kennedy, S. Nuclear RNAi maintains heritable gene silencing in Caenorhabditis elegans. Proc. Natl Acad. Sci. USA 108, 19683–19688 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. 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 

  97. Buckley, B. A. et al. A nuclear Argonaute promotes multigenerational epigenetic inheritance and germline immortality. Nature 489, 447–451 (2012). This study shows that HRDE-1 is the AGO protein responsible for transgenerational inheritance of classical RNAi-mediated silencing in C. elegans and that this involves H3K9 methylation of silenced genes in the germ line.

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Bagijn, M. P. et al. Function, targets, and evolution of Caenorhabditis elegans piRNAs. Science 337, 574–578 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Lee, H. C. et al. C. elegans piRNAs mediate the genome-wide surveillance of germline transcripts. Cell 150, 78–87 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Shirayama, M. et al. piRNAs initiate an epigenetic memory of nonself RNA in the C. elegans germline. Cell 150, 65–77 (2012). Together with references 96, 98 and 99, this paper defines the mechanism by which C. elegans piRNAs recognize and target non-self RNAs for silencing, a memory of which is then maintained by secondary siRNAs and the AGO protein HRDE-1.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Claycomb, J. M. et al. The Argonaute CSR-1 and its 22G-RNA cofactors are required for holocentric chromosome segregation. Cell 139, 123–134 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Seth, M. et al. The C. elegans CSR-1 argonaute pathway counteracts epigenetic silencing to promote germline gene expression. Dev. Cell 27, 656–663 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Wedeles, C. J., Wu, M. Z. & Claycomb, J. M. Protection of germline gene expression by the C. elegans Argonaute CSR-1. Dev. Cell 27, 664–671 (2013). References 102 and 103 demonstrate that the AGO protein CSR-1 protects its targets from repression by the PRG-1–HRDE-1 pathway. Thus, C. elegans uses small RNAs not only to mark non-self RNAs but also to designate expressed sequences as self.

    CAS  PubMed  Google Scholar 

  104. Conine, C. C. et al. Argonautes promote male fertility and provide a paternal memory of germline gene expression in C. elegans. Cell 155, 1532–1544 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Yang, L., Froberg, J. E. & Lee, J. T. Long noncoding RNAs: fresh perspectives into the RNA world. Trends Biochem. Sci. 39, 35–43 (2014).

    PubMed  Google Scholar 

  106. Ferrari, F., Alekseyenko, A. A., Park, P. J. & Kuroda, M. I. Transcriptional control of a whole chromosome: emerging models for dosage compensation. Nature Struct. Mol. Biol. 21, 118–125 (2014).

    CAS  Google Scholar 

  107. Kohlmaier, A. et al. A chromosomal memory triggered by Xist regulates histone methylation in X inactivation. PLoS Biol. 2, E171 (2004).

    PubMed  PubMed Central  Google Scholar 

  108. Plath, K. et al. Role of histone H3 lysine 27 methylation in X inactivation. Science 300, 131–135 (2003).

    CAS  PubMed  Google Scholar 

  109. Silva, J. et al. Establishment of histone H3 methylation on the inactive X chromosome requires transient recruitment of Eed–Enx1 polycomb group complexes. Dev. Cell 4, 481–495 (2003).

    CAS  PubMed  Google Scholar 

  110. Zhao, J., Sun, B. K., Erwin, J. A., Song, J. J. & Lee, J. T. Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science 322, 750–756 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Rinn, J. L. et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129, 1311–1323 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Pandey, R. R. et al. Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol. Cell 32, 232–246 (2008).

    CAS  PubMed  Google Scholar 

  113. Maenner, S. et al. 2D structure of the A region of Xist RNA and its implication for PRC2 association. PLoS Biol. 8, e1000276 (2010).

    PubMed  PubMed Central  Google Scholar 

  114. da Rocha, S. T. et al. Jarid2 is implicated in the initial Xist-induced targeting of PRC2 to the inactive X chromosome. Mol. Cell 53, 301–316 (2014).

    PubMed  Google Scholar 

  115. Brockdorff, N. Noncoding RNA and Polycomb recruitment. RNA 19, 429–442 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Davidovich, C., Zheng, L., Goodrich, K. J. & Cech, T. R. Promiscuous RNA binding by Polycomb repressive complex 2. Nature Struct. Mol. Biol. 20, 1250–1257 (2013).

    CAS  Google Scholar 

  117. Kaneko, S., Son, J., Shen, S. S., Reinberg, D. & Bonasio, R. PRC2 binds active promoters and contacts nascent RNAs in embryonic stem cells. Nature Struct. Mol. Biol. 20, 1258–1264 (2013).

    CAS  Google Scholar 

  118. Kanhere, A. et al. Short RNAs are transcribed from repressed Polycomb target genes and interact with Polycomb repressive complex-2. Mol. Cell 38, 675–688 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Zhao, J. et al. Genome-wide identification of Polycomb-associated RNAs by RIP-seq. Mol. Cell 40, 939–953 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Kaneko, S. et al. Interactions between JARID2 and noncoding RNAs regulate PRC2 recruitment to chromatin. Mol. Cell 53, 290–300 (2014). References 114 and 120 show that Jarid2 acts as an essential intermediate in Xist -mediated recruitment of PRC2 to chromatin, and that Xist repeat A is dispensable for PRC2 targeting.

    CAS  PubMed  Google Scholar 

  121. Kim, T. K. et al. Widespread transcription at neuronal activity-regulated enhancers. Nature 465, 182–187 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. De Santa, F. et al. A large fraction of extragenic RNA Pol II transcription sites overlap enhancers. PLoS Biol. 8, e1000384 (2010).

    PubMed  PubMed Central  Google Scholar 

  123. Orom, U. A. et al. Long noncoding RNAs with enhancer-like function in human cells. Cell 143, 46–58 (2010). This study reports the discovery of eRNAs, which are required to activate gene expression in cis.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Wang, K. C. et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 472, 120–124 (2011). This paper reports an eRNA that induces chromosomal looping and recruits H3K4 methylation to activate transcription of several genes in the HOXA cluster.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Lai, F. et al. Activating RNAs associate with Mediator to enhance chromatin architecture and transcription. Nature 494, 497–501 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Lam, M. T. et al. Rev–Erbs repress macrophage gene expression by inhibiting enhancer-directed transcription. Nature 498, 511–515 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Orom, U. A. & Shiekhattar, R. Long noncoding RNAs usher in a new era in the biology of enhancers. Cell 154, 1190–1193 (2013).

    PubMed  PubMed Central  Google Scholar 

  128. Harigaya, Y. et al. Selective elimination of messenger RNA prevents an incidence of untimely meiosis. Nature 442, 45–50 (2006). This study was the first to reveal that the mRNAs of meiosis-specific genes are targeted for destruction during vegetative growth in S. pombe . The authors defined a cis -RNA motif (the DSR) and a trans -protein factor (Mmi1) that are required for this regulation.

    CAS  PubMed  Google Scholar 

  129. Yamashita, A. et al. Hexanucleotide motifs mediate recruitment of the RNA elimination machinery to silent meiotic genes. Open Biol. 2, 120014 (2012).

    PubMed  PubMed Central  Google Scholar 

  130. Tashiro, S., Asano, T., Kanoh, J. & Ishikawa, F. Transcription-induced chromatin association of RNA surveillance factors mediates facultative heterochromatin formation in fission yeast. Genes Cells 18, 327–339 (2013).

    CAS  PubMed  Google Scholar 

  131. Zofall, M. et al. RNA elimination machinery targeting meiotic mRNAs promotes facultative heterochromatin formation. Science 335, 96–100 (2012). This paper shows that the machinery responsible for clearing meiotic mRNAs during vegetative growth also promotes the formation of heterochromatin islands at the corresponding meiotic gene loci.

    CAS  PubMed  Google Scholar 

  132. Sugiyama, T. & Sugioka-Sugiyama, R. Red1 promotes the elimination of meiosis-specific mRNAs in vegetatively growing fission yeast. EMBO J. 30, 1027–1039 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Lee, N. N. et al. Mtr4-like protein coordinates nuclear RNA processing for heterochromatin assembly and for telomere maintenance. Cell 155, 1061–1074 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Shah, S., Wittmann, S., Kilchert, C. & Vasiljeva, L. LncRNA recruits RNAi and the exosome to dynamically regulate pho1 expression in response to phosphate levels in fission yeast. Genes Dev. 28, 231–244 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Egan, E. D., Braun, C. R., Gygi, S. P. & Moazed, D. Post-transcriptional regulation of meiotic genes by a nuclear RNA silencing complex. RNA 20, 867–881 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Yamanaka, S., Yamashita, A., Harigaya, Y., Iwata, R. & Yamamoto, M. Importance of polyadenylation in the selective elimination of meiotic mRNAs in growing S. pombe cells. EMBO J. 29, 2173–2181 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Duret, L., Chureau, C., Samain, S., Weissenbach, J. & Avner, P. The Xist RNA gene evolved in eutherians by pseudogenization of a protein-coding gene. Science 312, 1653–1655 (2006).

    CAS  PubMed  Google Scholar 

  138. Gapp, K. et al. Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nature Neurosci. 17, 667–669 (2014).

    CAS  PubMed  Google Scholar 

  139. 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 

  140. Castel, S. E. & Martienssen, R. A. RNA interference in the nucleus: roles for small RNAs in transcription, epigenetics and beyond. Nature Rev. Genet. 14, 100–112 (2013).

    CAS  PubMed  Google Scholar 

  141. Bernstein, E., Caudy, A. A., Hammond, S. M. & Hannon, G. J. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409, 363–366 (2001).

    CAS  PubMed  Google Scholar 

  142. Elbashir, S. M., Lendeckel, W. & Tuschl, T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev. 15, 188–200 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Zamore, P. D., Tuschl, T., Sharp, P. A. & Bartel, D. P. RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell 101, 25–33 (2000).

    CAS  PubMed  Google Scholar 

  144. Parrish, S., Fleenor, J., Xu, S., Mello, C. & Fire, A. Functional anatomy of a dsRNA trigger: differential requirement for the two trigger strands in RNA interference. Mol. Cell 6, 1077–1087 (2000).

    CAS  PubMed  Google Scholar 

  145. Song, J. J. et al. The crystal structure of the Argonaute2 PAZ domain reveals an RNA binding motif in RNAi effector complexes. Nature Struct. Biol. 10, 1026–1032 (2003).

    CAS  PubMed  Google Scholar 

  146. Wang, Y. et al. Nucleation, propagation and cleavage of target RNAs in Ago silencing complexes. Nature 461, 754–761 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Ma, J. B., Ye, K. & Patel, D. J. Structural basis for overhang-specific small interfering RNA recognition by the PAZ domain. Nature 429, 318–322 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Ma, J. B. et al. Structural basis for 5′-end-specific recognition of guide RNA by the A. fulgidus Piwi protein. Nature 434, 666–670 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Song, J. J., Smith, S. K., Hannon, G. J. & Joshua-Tor, L. Crystal structure of Argonaute and its implications for RISC slicer activity. Science 305, 1434–1437 (2004).

    CAS  PubMed  Google Scholar 

  150. Matranga, C., Tomari, Y., Shin, C., Bartel, D. P. & Zamore, P. D. Passenger-strand cleavage facilitates assembly of siRNA into Ago2-containing RNAi enzyme complexes. Cell 123, 607–620 (2005).

    CAS  PubMed  Google Scholar 

  151. Rand, T. A., Petersen, S., Du, F. & Wang, X. Argonaute2 cleaves the anti-guide strand of siRNA during RISC activation. Cell 123, 621–629 (2005).

    CAS  PubMed  Google Scholar 

  152. Miyoshi, K., Tsukumo, H., Nagami, T., Siomi, H. & Siomi, M. C. Slicer function of Drosophila Argonautes and its involvement in RISC formation. Genes Dev. 19, 2837–2848 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Buker, S. M. et al. Two different Argonaute complexes are required for siRNA generation and heterochromatin assembly in fission yeast. Nature Struct. Mol. Biol. 14, 200–207 (2007).

    CAS  Google Scholar 

  154. Baulcombe, D. RNA silencing in plants. Nature 431, 356–363 (2004).

    CAS  PubMed  Google Scholar 

  155. Dalmay, T., Hamilton, A., Rudd, S., Angell, S. & Baulcombe, D. C. An RNA-dependent RNA polymerase gene in Arabidopsis is required for posttranscriptional gene silencing mediated by a transgene but not by a virus. Cell 101, 543–553 (2000).

    CAS  PubMed  Google Scholar 

  156. Sijen, T. et al. On the role of RNA amplification in dsRNA-triggered gene silencing. Cell 107, 465–476 (2001).

    CAS  PubMed  Google Scholar 

  157. Stuwe, E., Toth, K. F. & Aravin, A. A. Small but sturdy: small RNAs in cellular memory and epigenetics. Genes Dev. 28, 423–431 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Khurana, J. S. & Theurkauf, W. piRNAs, transposon silencing, and Drosophila germline development. J. Cell Biol. 191, 905–913 (2010).

    PubMed  PubMed Central  Google Scholar 

  159. Senti, K. A. & Brennecke, J. The piRNA pathway: a fly's perspective on the guardian of the genome. Trends Genet. 26, 499–509 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Ishizu, H., Nagao, A. & Siomi, H. Gatekeepers for Piwi–piRNA complexes to enter the nucleus. Curr. Opin. Genet. Dev. 21, 484–490 (2011).

    CAS  PubMed  Google Scholar 

  161. Brennecke, J. et al. Discrete small RNA-generating loci as master regulators of transposon activity in Drosophila. Cell 128, 1089–1103 (2007).

    CAS  PubMed  Google Scholar 

  162. Gunawardane, L. S. et al. A slicer-mediated mechanism for repeat-associated siRNA 5′ end formation in Drosophila. Science 315, 1587–1590 (2007). References 161 and 162 reported the discovery of piRNA amplification by the ping-pong mechanism.

    CAS  PubMed  Google Scholar 

  163. Halic, M. & Moazed, D. Dicer-independent primal RNAs trigger RNAi and heterochromatin formation. Cell 140, 504–516 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Yu, R., Jih, G., Iglesias, N. & Moazed, D. Determinants of heterochromatic siRNA biogenesis and function. Mol. Cell 53, 262–276 (2014).

    CAS  PubMed  Google Scholar 

  165. Kawakami, K., Hayashi, A., Nakayama, J. & Murakami, Y. A novel RNAi protein, Dsh1, assembles RNAi machinery on chromatin to amplify heterochromatic siRNA. Genes Dev. 26, 1811–1824 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Marasovic, M., Zocco, M. & Halic, M. Argonaute and Triman generate dicer-independent priRNAs and mature siRNAs to initiate heterochromatin formation. Mol. Cell 52, 173–183 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

D.H. was supported by the US National Science Foundation Graduate Research Fellowship Program. Research in D.M.'s laboratory is supported by grants from the US National Institutes of Health. D.M. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Danesh Moazed.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information S1 (figure)

Conservation and divergence of RNA silencing pathways. (PDF 205 kb)

PowerPoint slides

Glossary

RNA interference

(RNAi). Broadly refers to RNA silencing pathways that use Argonaute and PIWI proteins and small RNAs to silence gene expression.

Small interfering RNAs

(siRNAs). 22–24-nucleotide small RNAs that are generated from longer double-stranded RNA precursors by the ribonuclease Dicer.

Heterochromatin

Regions of chromatin that retain the condensed appearance of mitotic chromosomes throughout the cell cycle. Heterochromatic regions are associated with repressive histone modifications and structural proteins, and are transcriptionally silent.

Argonaute

(AGO). A family of proteins that bind to small RNAs and that are conserved in all domains of life. They mediate target recognition via base-pairing interactions between their bound small RNA and complementary coding or non-coding RNAs.

Heterochromatic gene silencing

Silencing of gene expression within heterochromatin. It was originally thought to exclusively involve transcriptional gene silencing mechanisms, but recent findings indicate that co-transcriptional degradation of nascent RNA, or co-transcriptional gene silencing, also play important parts in silencing.

Pericentromeric DNA repeat

A repeated DNA sequence that surrounds the centromeres of most eukaryotic chromosomes. These repeats are assembled into heterochromatin, which has been demonstrated to have roles in cohesin recruitment in fission yeast and mammals, and de novo centromere assembly in fission yeast.

RNA-induced transcriptional silencing

(RITS). A protein complex first identified in Schizosaccharomyces pombe. In addition to Argonaute 1, the RITS complex contains a GW domain protein, Tas3, and a chromodomain protein, Chp1, which tether the complex to the chromosome via interactions with nascent long non-coding RNAs and nucleosomes with methylated histone H3 lysine 9.

Long terminal repeat

(LTR). A DNA sequence that is repeated at the ends of retrotransposons or pro-viral DNA that is formed from retroviral RNA by reverse transcription. Plant and mammalian genomes contain thousands of LTRs.

Epigenetic phenomena

Phenomena in which changes in gene expression occur without a corresponding change in the DNA sequence; such changes are stable in the absence of initiating signals.

Paramutation

The ability of a silent allele to convert an active allele to the silent (and paramutagenic) form. It was first described in Zea mays.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Holoch, D., Moazed, D. RNA-mediated epigenetic regulation of gene expression. Nat Rev Genet 16, 71–84 (2015). https://doi.org/10.1038/nrg3863

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3863

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