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.

  • Article
  • Published:

LSM2-8 and XRN-2 contribute to the silencing of H3K27me3-marked genes through targeted RNA decay

Abstract

In fission yeast and plants, RNA processing and degradation contribute to heterochromatin silencing, alongside conserved pathways of transcriptional repression. It has not been known whether similar pathways exist in metazoans. Here, we describe a pathway of silencing in Caenorhabditis elegans somatic cells, in which the highly conserved RNA-binding complex LSM2-8 contributes selectively to the repression of heterochromatic reporters and endogenous genes bearing the Polycomb mark, histone H3K27me3. This acts by degrading selected transcripts through the XRN-2 exoribonuclease. Disruption of the LSM2-8 pathway leads to mRNA stabilization. Unlike previously described pathways of heterochromatic RNA degradation, LSM2-8-mediated RNA degradation does not target nor require H3K9 methylation. Intriguingly, loss of this pathway coincides with a localized reduction in H3K27me3 at lsm-8-sensitive loci. We have thus uncovered a mechanism of RNA degradation that selectively contributes to the silencing of a subset of H3K27me3-marked genes, revealing a previously unrecognized layer of post-transcriptional control in metazoan heterochromatin.

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

Fig. 1: LSM proteins silence heterochromatic reporters, but not euchromatic reporters.
Fig. 2: LSM2-8 mediates heterochromatic silencing and prevents sterility and premature death.
Fig. 3: The LSM2-8 complex silences endogenous transcripts and acts both independently and additively to H3K9 methylation.
Fig. 4: Over 93% of genes silenced by LSM2-8 carry the Polycomb mark H3K27me3.
Fig. 5: lsm-7, like the EZH2 homologue mes-2, is required to silence the egl-5 Hox gene.
Fig. 6: LSM2-8 and XRN-2 work on the same silencing pathway.
Fig. 7: LSM2-8 mediates silencing primarily through RNA degradation.

Similar content being viewed by others

Data availability

RNA-seq data that support the findings of this study have been deposited in the Gene Expression Omnibus (GEO) under accession code GSE92851. Previously published ChIP-seq data that were re-analysed here are available from ModEncode (http://data.modencode.org/) for L3_H3K9me1/2/3 (5036, 5050, 5037, 5040), L3_H3K27me3 (5045, 5051), L3_H3K27ac (5054), L3_H3K4me2/3 (5055, 3576). Source data for Figs. 1–7 and Extended Data Figs. 1, 2 and 48 are presented with the paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Saksouk, N., Simboeck, E. & Dejardin, J. Constitutive heterochromatin formation and transcription in mammals. Epigenetics Chromatin 8, 3 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Trojer, P. & Reinberg, D. Facultative heterochromatin: is there a distinctive molecular signature? Mol. Cell 28, 1–13 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Wenzel, D., Palladino, F. & Jedrusik-Bode, M. Epigenetics in C. elegans: facts and challenges. Genesis 49, 647–661 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. Zeller, P. et al. Histone H3K9 methylation is dispensable for Caenorhabditis elegans development but suppresses RNA:DNA hybrid-associated repeat instability. Nat. Genet. 48, 1385–1395 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Gaydos, L. J., Wang, W. & Strome, S. Gene repression. H3K27me and PRC2 transmit a memory of repression across generations and during development. Science 345, 1515–1518 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wang, J., Jia, S. T. & Jia, S. New insights into the regulation of heterochromatin. Trends Genet. 32, 284–294 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Buhler, M. RNA turnover and chromatin-dependent gene silencing. Chromosoma 118, 141–151 (2009).

    Article  CAS  PubMed  Google Scholar 

  8. Grewal, S. I. & Elgin, S. C. Transcription and RNA interference in the formation of heterochromatin. Nature 447, 399–406 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. 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).

    Article  CAS  PubMed  Google Scholar 

  12. Eberle, A. B. et al. An interaction between RRP6 and SU(VAR)3-9 Targets RRP6 to Heterochromatin and contributes to heterochromatin maintenance in Drosophila melanogaster. PLoS Genet. 11, e1005523 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Shin, J. H. et al. The role of the Arabidopsis exosome in siRNA-independent silencing of heterochromatic loci. PLoS Genet. 9, e1003411 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Vasiljeva, L., Kim, M., Terzi, N., Soares, L. M. & Buratowski, S. Transcription termination and RNA degradation contribute to silencing of RNA polymerase II transcription within heterochromatin. Mol. Cell 29, 313–323 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Keller, C. et al. HP1(Swi6) mediates the recognition and destruction of heterochromatic RNA transcripts. Mol. Cell 47, 215–227 (2012).

    Article  CAS  PubMed  Google Scholar 

  16. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zofall, M. et al. RNA elimination machinery targeting meiotic mRNAs promotes facultative heterochromatin formation. Science 335, 96–100 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Yamanaka, S. et al. RNAi triggered by specialized machinery silences developmental genes and retrotransposons. Nature 493, 557–560 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Touat-Todeschini, L. et al. Selective termination of lncRNA transcription promotes heterochromatin silencing and cell differentiation. EMBO J. 36, 2626–2641 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. St-Andre, O. et al. Negative regulation of meiotic gene expression by the nuclear poly(A)-binding protein in fission yeast. J. Biol. Chem. 285, 27859–27868 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tucker, J. F. et al. A novel epigenetic silencing pathway involving the highly conserved 5′-3′ exoribonuclease Dhp1/Rat1/Xrn2 in Schizosaccharomyces pombe. PLoS Genet. 12, e1005873 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Chalamcharla, V. R., Folco, H. D., Dhakshnamoorthy, J. & Grewal, S. I. Conserved factor Dhp1/Rat1/Xrn2 triggers premature transcription termination and nucleates heterochromatin to promote gene silencing. Proc. Natl Acad. Sci. USA 112, 15548–15555 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang, X. et al. Targeting of Polycomb repressive complex 2 to RNA by short repeats of consecutive guanines. Mol. Cell 65, 1056–1067 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Towbin, B. D. et al. Step-wise methylation of histone H3K9 positions heterochromatin at the nuclear periphery. Cell 150, 934–947 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Tharun, S. Roles of eukaryotic Lsm proteins in the regulation of mRNA function. Int. Rev. Cell Mol. Biol. 272, 149–189 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Beggs, J. D. Lsm proteins and RNA processing. Biochem. Soc. Trans. 33, 433–438 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Kufel, J., Bousquet-Antonelli, C., Beggs, J. D. & Tollervey, D. Nuclear pre-mRNA decapping and 5′ degradation in yeast require the Lsm2-8p complex. Mol. Cell. Biol. 24, 9646–9657 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Golisz, A., Sikorski, P. J., Kruszka, K. & Kufel, J. Arabidopsis thaliana LSM proteins function in mRNA splicing and degradation. Nucleic Acids Res. 41, 6232–6249 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Perea-Resa, C., Hernandez-Verdeja, T., Lopez-Cobollo, R., del Mar Castellano, M. & Salinas, J. LSM proteins provide accurate splicing and decay of selected transcripts to ensure normal Arabidopsis development. Plant Cell 24, 4930–4947 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cornes, E. et al. Cytoplasmic LSM-1 protein regulates stress responses through the insulin/IGF-1 signaling pathway in Caenorhabditis elegans. RNA 21, 1544–1553 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Meister, P., Towbin, B. D., Pike, B. L., Ponti, A. & Gasser, S. M. The spatial dynamics of tissue-specific promoters during C. elegans development. Genes Dev. 24, 766–782 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Towbin, B. D., Meister, P., Pike, B. L. & Gasser, S. M. Repetitive transgenes in C. elegans accumulate heterochromatic marks and are sequestered at the nuclear envelope in a copy-number- and lamin-dependent manner. Cold Spring Harbor Symp. Quant. Biol. 75, 555–565 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hendriks, G. J., Gaidatzis, D., Aeschimann, F. & Grosshans, H. Extensive oscillatory gene expression during C. elegans larval development. Mol. Cell 53, 380–392 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Ho, J. W. et al. Comparative analysis of metazoan chromatin organization. Nature 512, 449–452 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, T. et al. Broad chromosomal domains of histone modification patterns in C. elegans. Genome Res. 21, 227–236 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ahringer, J. & Gasser, S. M. Repressive chromatin in Caenorhabditis elegans: establishment, composition and function. Genetics 208, 491–511 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Conway, E., Healy, E. & Bracken, A. P. PRC2 mediated H3K27 methylations in cellular identity and cancer. Curr. Opin. Cell Biol. 37, 42–48 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Grossniklaus, U. & Paro, R. Transcriptional silencing by polycomb-group proteins. Cold Spring Harb. Perspect. Biol. 6, a019331 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Patel, T., Tursun, B., Rahe, D. P. & Hobert, O. Removal of Polycomb repressive complex 2 makes C. elegans germ cells susceptible to direct conversion into specific somatic cell types. Cell Rep. 2, 1178–1186 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ketel, C. S. et al. Subunit contributions to histone methyltransferase activities of fly and worm polycomb group complexes. Mol. Cell. Biol. 25, 6857–6868 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yuzyuk, T., Fakhouri, T. H., Kiefer, J. & Mango, S. E. The polycomb complex protein mes-2/E(z) promotes the transition from developmental plasticity to differentiation in C. elegans embryos. Dev. Cell 16, 699–710 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ross, J. M. & Zarkower, D. Polycomb group regulation of Hox gene expression in C. elegans. Dev. Cell 4, 891–901 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Hench, J. et al. The Homeobox genes of Caenorhabditis elegans and insights into their spatio-temporal expression dynamics during embryogenesis. PLoS One 10, e0126947 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Ferreira, H. B., Zhang, Y., Zhao, C. & Emmons, S. W. Patterning of Caenorhabditis elegans posterior structures by the Abdominal-B homolog, egl-5. Dev. Biol. 207, 215–228 (1999).

    Article  CAS  PubMed  Google Scholar 

  48. Zhou, L. et al. Crystal structures of the Lsm complex bound to the 3′ end sequence of U6 small nuclear RNA. Nature 506, 116–120 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Miki, T. S., Carl, S. H., Stadler, M. B. & Grosshans, H. XRN2 autoregulation and control of polycistronic gene expresssion in Caenorhabditis elegans. PLoS Genet. 12, e1006313 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Miki, T. S. & Grosshans, H. The multifunctional RNase XRN2. Biochem. Soc. Trans. 41, 825–830 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Miki, T. S., Ruegger, S., Gaidatzis, D., Stadler, M. B. & Grosshans, H. Engineering of a conditional allele reveals multiple roles of XRN2 in Caenorhabditis elegans development and substrate specificity in microRNA turnover. Nucleic Acids Res. 42, 4056–4067 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lotan, R., Goler-Baron, V., Duek, L., Haimovich, G. & Choder, M. The Rpb7p subunit of yeast RNA polymerase II plays roles in the two major cytoplasmic mRNA decay mechanisms. J. Cell Biol. 178, 1133–1143 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Haimovich, G., Choder, M., Singer, R. H. & Trcek, T. The fate of the messenger is pre-determined: a new model for regulation of gene expression. Biochim. Biophys. Acta 1829, 643–653 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Coy, S. & Vasiljeva, L. The exosome and heterochromatin: multilevel regulation of gene silencing. Adv. Exp. Med. Biol. 702, 105–121 (2011).

    Article  PubMed  Google Scholar 

  55. Hurschler, B. A., Harris, D. T. & Grosshans, H. The type II poly(A)-binding protein PABP-2 genetically interacts with the let-7 miRNA and elicits heterochronic phenotypes in Caenorhabditis elegans. Nucleic Acids Res. 39, 5647–5657 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kuhn, U. et al. Poly(A) tail length is controlled by the nuclear poly(A)-binding protein regulating the interaction between poly(A) polymerase and the cleavage and polyadenylation specificity factor. J. Biol. Chem. 284, 22803–22814 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Lemieux, C. & Bachand, F. Cotranscriptional recruitment of the nuclear poly(A)-binding protein Pab2 to nascent transcripts and association with translating mRNPs. Nucleic Acids Res. 37, 3418–3430 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Beaulieu, Y. B., Kleinman, C. L., Landry-Voyer, A. M., Majewski, J. & Bachand, F. Polyadenylation-dependent control of long noncoding RNA expression by the poly(A)-binding protein nuclear 1. PLoS Genet. 8, e1003078 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Soshnikova, N. & Duboule, D. Epigenetic temporal control of mouse Hox genes in vivo. Science 324, 1320–1323 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Bender, L. B., Cao, R., Zhang, Y. & Strome, S. The MES-2/MES-3/MES-6 complex and regulation of histone H3 methylation in C. elegans. Curr. Biol. 14, 1639–1643 (2004).

    Article  CAS  PubMed  Google Scholar 

  61. Zhang, H. et al. The C. elegans Polycomb gene SOP-2 encodes an RNA binding protein. Mol. Cell 14, 841–847 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Zhang, T. et al. RNA-binding proteins SOP-2 and SOR-1 form a novel PcG-like complex in C. elegans. Development 133, 1023–1033 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Djupedal, I. et al. RNA Pol II subunit Rpb7 promotes centromeric transcription and RNAi-directed chromatin silencing. Genes Dev. 19, 2301–2306 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ringrose, L. Noncoding RNAs in polycomb and trithorax regulation: a quantitative perspective. Annu. Rev. Genet. 51, 385–411 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Johnson, W. L. & Straight, A. F. RNA-mediated regulation of heterochromatin. Curr. Opin. Cell Biol. 46, 102–109 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wang, X. & Paucek, R. D. Molecular analysis of PRC2 recruitment to DNA in chromatin and its inhibition by RNA. Nat. Struct. Mol. Biol. 24, 1028–1038 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Katic, I., Xu, L. & Ciosk, R. CRISPR/Cas9 genome editing in Caenorhabditis elegans: evaluation of templates for homology-mediated repair and knock-ins by homology-independent DNA repair. G3 (Bethesda) 5, 1649–1656 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Dickinson, D. J., Ward, J. D., Reiner, D. J. & Goldstein, B. Engineering the Caenorhabditis elegans genome using Cas9-triggered homologous recombination. Nat. Methods 10, 1028–1034 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Timmons, L., Court, D. L. & Fire, A. Ingestion of bacterially expressed dsRNAs can produce specific and potent genetic interference in Caenorhabditis elegans. Gene 263, 103–112 (2001).

    Article  CAS  PubMed  Google Scholar 

  71. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Au, K. F., Jiang, H., Lin, L., Xing, Y. & Wong, W. H. Detection of splice junctions from paired-end RNA-seq data by SpliceMap. Nucleic Acids Res. 38, 4570–4578 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Habacher, C. et al. Ribonuclease-mediated control of body fat. Dev. Cell 39, 359–369 (2016).

    Article  CAS  PubMed  Google Scholar 

  74. Latorre, I. et al. The DREAM complex promotes gene body H2A.Z for target repression. Genes Dev. 29, 495–500 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Wang, J. J. et al. The influences of PRG-1 on the expression of small RNAs and mRNAs. BMC Genomics 15, 321 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

The accession number for the RNA-seq data in the NCBI Gene Expression Omnibus is GSE92851. Some strains were provided by the Caenorhabditis Genetics Center (CGC), which is funded by NIH Office of Research Infrastructure Programs (P40 OD010440). We thank I. Katiç, the FMI Genomics and Microscopy facilities, P. Zeller and M. Fukushima for technical help, advice and discussions, and T.S. Miki for access to data and discussions. We thank M. Bühler, H. Großhans and W. Filipowicz for discussions and proofreading of the text. We acknowledge support from the Novartis Research Foundation, as well as a Marie Curie Intra-European grant (#PIEF-GA-2010-276589) and Swiss National Science Foundation Marie-Heim Vögtlin grant (#PMPDP3_151381, # PMPDP3_168717) to A.M., an SNF grant (#310030B_156936) to S.M.G. and support from NCCR RNA & Disease to H. Großhans (to F.A.). This project received funding from the European Research Council under the European Union’s Horizon 2020 Research and Innovation programme (Epiherigans; grant no. 743312 to S.M.G.).

Author information

Authors and Affiliations

Authors

Contributions

A.M. planned and executed most experiments, evaluated results and wrote the paper. S.M.G. planned experiments, evaluated results and wrote the paper. D.G., with A.M., analysed the RNA-seq data and other genome-wide data. C.S. analysed the L1 RNA-seq data. J.P. performed the H3K27me3 ChIP-qPCR experiment and analysis. V.K. performed the gonad staining and analysis. F.A. helped generate the lsm-8 mutant strain.

Corresponding author

Correspondence to Susan M. Gasser.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 LSM proteins are highly conserved and silence heterochromatic, but not euchromatic reporters.

a, LSM protein comparison between C. elegans and H. sapiens. b, Heterochromatic reporters derepression at all developmental stages. lsm-7 RNAi is compared to control RNAi. Derepression monitored by GFP live imaging was assessed at the embryonic stage (strain GW566, Supplementary Table 1, Bar: 10 μm), with nuclei enlarged in the inset and at larval stages L1-L4, (strain GW306, Supplementary Table 1, Bar: 50 μm, Bar: 100 μm for gravid adults). These observations were repeated ten times independently with similar results. c, Quantitation of derepression assays. In L1 progeny under gut-2/lsm-2, lsm-5, lsm-6 and control RNAi conditions (mock: negative control and mes-4: positive control), the GFP fluorescence intensity of the heterochromatic reporter pkIS1582 was measured by the worm sorter. F2: second generation. Quantification and statistical analysis were based on n =375 worms for each condition pooled from three independent experiments. Data are displayed as in Fig. 1e. P values indicated were calculated with a two-tailed unpaired t test. d, Quantitation of derepression of different heterochromatic reporters (Supplementary Table 1). P values indicated were calculated with a two- tailed unpaired t test. Quantification and statistical analysis were based on n= 1460, 2399, 2631, 3850, 634, 1855 worms for conditions indicated from left to right, pooled from two independent experiments. e, Confirmation of lsm-1 and lsm-7 knockdown by RNAi. qPCR analysis of lsm-7 and lsm-1 mRNA in L1 worms upon mock, lsm-7 or lsm-1 RNAi treatments. lsm-7 and lsm-1 mRNA are expressed relative to the levels in mock RNAi condition. Bars represent mean value derived from three (lsm-7 RNAi) and two independent experiments (lsm-1 RNAi), with the value of each experiment shown as dots. f, Quantitation of fluorescence intensity of the euchromatic reporter (GW849, gain2) in L1 progeny. P values calculated as in (c). Quantification and statistical analysis were based on n =375 worms for each condition pooled from three independent experiments. g, Same as in (f), with a gain=1 for the fluorescence of both the heterochromatic (GW306) and euchromatic (GW849) reporters. P values as in (c). Quantification and statistical analysis were based on n =370 worms for each condition, pooled from two independent experiments. Statistical source data are provided in Source Data. Extended Data Fig. 1.

Source data

Extended Data Fig. 2 lsm-8-/- mutant worms are 100% sterile but developing gonads resemble WT through L3 and L4 stages.

a, Z-projection of confocal images showing fixed DAPI staining of a WT (N2) worm, at L3 stage. Gonad arms are highlighted by the red dashed line and same to right with a lsm-8-/- L3 larva (GW1125). b, Quantification of the length, width and gonad nuclei count. Bars represent mean value derived from two independent experiments, with the value of each experiment shown as the dot. The two experiments examined 4 worms in total per genotype. c, DIC image of a WT young adult (YA) with a normal anatomy and normal gonad (red dashed line) with oocytes (pink shading). The white arrow indicates the vulva as in YA. DIC image of lsm-8-/-, lsm-2-/- and lsm- 5-/- YA. The gonad (red dashed line) has no forming oocytes and has an abnormal composition of cells at that stage. Black arrows indicate the presence of vacuoles. d, Heterochromatic reporter (pkIs1582) derepression in WT (GW306) background following lsm-7 RNAi in a L4 larva. The enlargement to the right shows the gonad (red dashed line) with germ cells which are not derepressed. e, Merge DIC and live GFP microscopy of lsm-8 mutant (GW1119) carrying the heterochromatic reporter pkIs1582, at the L4 larvae stage as confirmed by the vulva in the inset. The derepression of the reporter in the gonad is not detectable in germ cells, but is in the somatic gonad cells marked with asterisks: DTC (distal tip cells), gonadal sheath, spermathecal cells. f, Z-projection of confocal images showing the nuclear GFP derepression of the heterochromatic reporter pkIs1582 (GW1119) in nearly all if not all somatic cells of an lsm8-/- worm. g, GFP and DIC merged images at a single focal plan showing the optimal view of germ cells (inside dashed red line), which are not derepressed in lsm8-/- worm (GW1119) even treated with RNAi against piRNA factors such as csr-1. Scale Bar in a, c-g, 50 μm. Data in a and c-g represent results from three independent experiments, except for a and g, where the experiments have been performed twice with similar results. Statistical source data are provided in Source Data Extended Data Fig. 2.

Source data

Extended Data Fig. 3 Worm sorting and quantification settings based on gating region.

a, COPAS Biosort conditions optimised for the quantification of the heterochromatic reporter fluorescence. The COPAS Biosort (Union Biometrica) machine is an adapted flow cytometry version that can be used in order to quantify and collect worms according to their size and fluorescence criteria. The upper panel reflects the gating region (black diamond) based on the optical density of the object (optical extinction, EXT in the y axis) and the axial length (time of flight, TOF, in the x axis) of the object selecting the L1 worm population, as determined empirically in pilot experiments by verifying the stage through microscopic examination of sorted worms with this gate criteria. The same criteria gating was identical for every quantification of the heterochromatic reporter fluorescence. The lower panel shows the worm distribution of the size-selected worms based on green parameters (green peak height (green PH) and green peak width (green PW). This lower panel in a, represents the fluorescence of the heterochromatic reporter (GW306) in control RNAi condition and in b, in lsm-7 RNAi conditions. c, COPAS Biosort conditions optimised for the sorting of homozygous lsm-8 mutant at the L3 stage. The upper panel reflects the gating region (black diamond) based on the optical density of the objects (optical extinction, EXT in the y axis) and the axial length (Time of Flight, TOF, in the x axis) of the objects selecting the L3 worm population. The lower panel shows the worm distribution based on green parameters (green peak height (green PH) and green peak width (green PW)), the second gating region (black window) shown in that panel selects here the non-green worms, homozygous for lsm-8. The gating strategies were determined empirically in pilot experiments by verifying the size, shape gonad and vulva developmental stage by microscopic examination. Morphological validations during the sorting process were also performed. Sorting of the homozygous animals was done by selecting non- GFP pharynx animals, and the gating was also determined stringently by examining the two populations and by verifying the different criteria with fluorescent microscopy.

Extended Data Fig. 4 Control of the developmental timing of the RNA-seq samples within L3 stage larvae.

a, Gene expression data were collected over larval development at 25 °C and the average expression of somatic genes that were found to increase during this time course (rising somatic genes) is plotted in the left part ([35], see Methods). This analysis allowed us to compare the average expression of somatic genes that increase naturally during development to the average expression in our RNA-seq samples within the L3 larval stage. Samples from the four biological replicates of the four different genotypes that were the closest by developmental timing were selected accordingly and assigned to replica 1 and replica 2. Those two matched replicates (developmentally timed) were used for the main bioinformatics analysis, but the other samples were used for additional validations, and confirmed the main findings. b, Relative gene expression profiles as scatter plots. Fold-change (log2) in gene expression of two biological replicas of RNA-seq from sorted L3 worms of lsm-8-/-, met-2-/- set-25-/- and the triple (lsm-8-/-, met-2-/- set-25-/-) mutant versus WT. Each dot corresponds to a gene. Red dots here are rising genes, genes with increased expression level during the time course described [35], which do not change significantly in any of the mutant strains. Statistical source data are provided in Source Data Extended Data Fig. 4.

Source data

Extended Data Fig. 5 Genes silenced by LSM2-8 have a low steady-state expression and are not enriched on chromosome arms.

a, Scatter plots comparing the mean of log2(Fc) in lsm-8-/- vs WT (x axis) from two independent RNA-seq with the H3K9me1 mark ChIP-seq data and with additional ChIP- seq data for H3K9me3 and H3K27me3 (y axis) from ModEncode using different antibodies than those used in Figure 4. b, Scatter plots comparing absolute transcript abundances (log2 of normalized reads count) of annotated genes in lsm-8-/-, met-2-/- set-25-/- and the triple (lsm-8-/-, met-2-/- set-25-/-) mutant versus WT from the two biological replica 1 and 2. Boxes with pink background indicate low abundance values smaller than 6 in log2 scale for genes considered to be repressed in WT. This corresponds to <64 normalized RNA-seq reads per gene, in contrast to 1024 reads per gene represented by a value of 10. Note the large proportion among the genes upregulated in the assessed mutants (above the diagonal), which are repressed or very poorly expressed in WT. c, Distribution of upregulated genes in lsm-8-/- along chromosomes. LEM-2 ChIP enrichment plotted over chromosomes (embryonic WT data from [76] is in grey, indicating proximity to the nuclear periphery. Up- regulated genes in lsm-8-/- (FDR <0.05 and Fc >4) represented by the red dots are plotted over autosomes and X chromosome. Data shown represent two RNA-seq experiments. d, Comparison between our RNA-seq and other available RNA-seq datasets [49, 74, 75] in L3 stage C. elegans, for the percentage of H3K27me3-enriched genes among misregulated genes. Average of two replicas. We classify a gene as enriched for H3K27me3, if it has positive reproducible enrichment of H3K27me3 over input from two ChIP-seq datasets from ModEncode (Supplementary Table 3). Genes upregulated in xrn-2 RNAi treated worms [49] but not upregulated in lsm-8 mutant worms are not significantly enriched for H3K27me3 (Supplementary Table 2). Statistical source data are provided in Source Data Extended Data Fig. 5.

Source data

Extended Data Fig. 6 LSM-8 ablation does not alter transcription termination accuracy, strand specificity nor splicing.

a, UCSC genome browser view showing wiggle tracks from positive (+) or negative (-) strands show the differential expression of the col-2 gene, which is upregulated in lsm-8-/- compared to WT (y axis in log2). Data shown are derived from the two independent biological RNA-seq replicas. The expression level of the neighboring genes is not affected and termination defects are not observed. All introns were as efficiently spliced in lsm-8-/- as in WT. b, G browse view showing the ModEncode ChIP-seq tracks for H3K27me1, H3K27me3 (two different antibodies) and H3K27Ac at the same genomic locus (IV:10,082,495..10, 087, 496) around the col-2 gene, as shown in (a). The col-2 gene is upregulated in lsm-8-/- compared to WT and enriched for H3K27me3, as 95% of the genes upregulated in lsm-8-/-. Statistical source data are provided in Source Data Extended Data Fig. 6.

Source data

Extended Data Fig. 7 lsm-8 deletion does not affect splicing globally.

a, RNA IP-qPCR. LSM-4-FLAG RNA IP analysis in native conditions. RNA levels were normalized to input and U1snRNA levels. ZK970.7 is upregulated in lsm-8-/- (lsm-8 target gene) and associate with LSM4 (>1), whereas F08G2.8 is not (non-target gene) and do not associate with LSM-4. Those two examples suggest that the LSM-8 complex can bind to the RNAs it regulates. Bars represent mean value derived from two independent experiments, with the value of each experiment shown as a dot. b, Reads which align on exon-exon junctions were counted in lsm-8-/- and WT worms. Scatter plot compares exon-exon junction mapped reads (log2) normalized to their intrinsic gene level in WT (x-axis) and lsm-8-/- worms (y-axis). r: Pearson correlation coefficient. c, List of genes including the 18 exon-exon junctions reproducibly affected in lsm-8-/- worms as in (b). Statistical source data are provided in Source Data Extended Data Fig. 7.

Source data

Extended Data Fig. 8 LSM2-8 promotes the degradation of specific transcripts.

a, Scheme of the RNA decay assay. WT and lsm-8 -/- worms were sorted, re-fed with OP50 in liquid culture for 1h at room temperature and treated with 50 μg/ml final concentration of α-amanitin, which inhibits Pol II and Pol III transcription. RNA was isolated at time 0, 4.5h and at 6h, as indicated for each independent experiment. b, RNA levels of three transcripts affected by LSM-8 (upper graph) and two control transcripts (expression not affected by LSM-8, lower graph) were determined by RT- qPCR and normalized to 18S rRNA levels which are insensitive to α-amanitin. The value at 0h is defined as 100%. Bars represent mean value derived from four independent experiments for eft-3, from three independent experiments for far-3, ZK970.7 and F08G2.8 and two independent experiments for grl-23, with the value of each experiment shown as the dot. Statistical source data are provided in Source Data Extended Data Fig. 8.

Source data

Supplementary information

Supplementary Information

Supplementary Note and reference associated with Extended Data Figures.

Reporting Summary

Supplementary Table 1

List of strains and primers used in this study.

Supplementary Table 2

Processed RNA-seq data for genes in lsm-8, met-2set-25 and lsm-8, met-2set-25 triple mutant versus WT at the L3 stage.

Supplementary Table 3

Enrichment of histone marks in (ModEncode Data)

Supplementary Table 4

Processed RNA-seq data for genes in lsm-8 mutant versus WT at the L1 stage.

Supplementary Table 5

Lists of biological processes affected in the different mutants, GO analysis.

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 2

Statistical source data

Source Data Fig. 3

Statistical source data

Source Data Fig. 4

Statistical source data

Source Data Fig. 5

Statistical source data

Source Data Fig. 6

Statistical source data

Source Data Fig. 7

Statistical source data

Source Data Extended Data Fig. 1

Statistical source data

Source Data Extended Data Fig. 2

Statistical source data

Source Data Extended Data Fig. 4

Statistical source data

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 7

Statistical source data

Source Data Extended Data Fig. 8

Statistical source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mattout, A., Gaidatzis, D., Padeken, J. et al. LSM2-8 and XRN-2 contribute to the silencing of H3K27me3-marked genes through targeted RNA decay. Nat Cell Biol 22, 579–590 (2020). https://doi.org/10.1038/s41556-020-0504-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-0504-1

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