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:

Regulation of gene expression by cis-acting long non-coding RNAs

Abstract

Long non-coding RNAs (lncRNAs) are diverse transcription products emanating from thousands of loci in mammalian genomes. Cis-acting lncRNAs, which constitute a substantial fraction of lncRNAs with an attributed function, regulate gene expression in a manner dependent on the location of their own sites of transcription, at varying distances from their targets in the linear genome. Through various mechanisms, cis-acting lncRNAs have been demonstrated to activate, repress or otherwise modulate the expression of target genes. We discuss the activities that have been ascribed to cis-acting lncRNAs, the evidence and hypotheses regarding their modes of action, and the methodological advances that enable their identification and characterization. The emerging principles highlight lncRNAs as transcriptional units highly adept at contributing to gene regulatory networks and to the generation of fine-tuned spatial and temporal gene expression programmes.

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: Genomic locations and prevalence of cis-acting long non-coding RNAs.
Fig. 2: Mechanisms of action of activating cis-acting long non-coding RNAs.
Fig. 3: Functions of repressive cis-acting long non-coding RNAs.
Fig. 4: Plausible functions of cis-acting long non-coding RNAs.
Fig. 5: Methods for mapping functional features in cis-acting long non-coding RNAs.

Similar content being viewed by others

References

  1. Guttman, M. et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458, 223–227 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Mercer, T. R. et al. Targeted RNA sequencing reveals the deep complexity of the human transcriptome. Nat. Biotechnol. 30, 99–104 (2011).

    PubMed  PubMed Central  Google Scholar 

  3. Carninci, P. et al. The transcriptional landscape of the mammalian genome. Science 309, 1559–1563 (2005).

    CAS  PubMed  Google Scholar 

  4. Samata, M. & Akhtar, A. Dosage compensation of the X chromosome: a complex epigenetic assignment involving chromatin regulators and long noncoding RNAs. Annu. Rev. Biochem. 87, 323–350 (2018).

    CAS  PubMed  Google Scholar 

  5. Sherstyuk, V. V., Medvedev, S. P. & Zakian, S. M. Noncoding RNAs in the regulation of pluripotency and reprogramming. Stem Cell Rev. 14, 58–70 (2018).

    CAS  Google Scholar 

  6. Quinodoz, S. & Guttman, M. Long noncoding RNAs: an emerging link between gene regulation and nuclear organization. Trends Cell Biol. 24, 651–663 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Lin, C. & Yang, L. Long noncoding RNA in cancer: wiring signaling circuitry. Trends Cell Biol. 28, 287–301 (2018).

    CAS  PubMed  Google Scholar 

  8. Scheuermann, J. C. & Boyer, L. A. Getting to the heart of the matter: long non-coding RNAs in cardiac development and disease. EMBO J. 32, 1805–1816 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Guo, J., Liu, Z. & Gong, R. Long noncoding RNA: an emerging player in diabetes and diabetic kidney disease. Clin. Sci. 133, 1321–1339 (2019).

    CAS  Google Scholar 

  10. Zuckerman, B. & Ulitsky, I. Predictive models of subcellular localization of long RNAs. RNA 25, 557–572 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Tilgner, H. et al. Deep sequencing of subcellular RNA fractions shows splicing to be predominantly co-transcriptional in the human genome but inefficient for lncRNAs. Genome Res. 22, 1616–1625 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Melé, M. et al. Chromatin environment, transcriptional regulation, and splicing distinguish lincRNAs and mRNAs. Genome Res. 27, 27–37 (2017).

    PubMed  PubMed Central  Google Scholar 

  13. Hezroni, H. et al. Principles of long noncoding RNA evolution derived from direct comparison of transcriptomes in 17 species. Cell Rep. 11, 1110–1122 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Tichon, A. et al. A conserved abundant cytoplasmic long noncoding RNA modulates repression by Pumilio proteins in human cells. Nat. Commun. 7, 12209 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Lee, S. et al. Noncoding RNA NORAD regulates genomic stability by sequestering PUMILIO proteins. Cell 164, 69–80 (2016).

    CAS  PubMed  Google Scholar 

  16. Musgrove, C., Jansson, L. I. & Stone, M. D. New perspectives on telomerase RNA structure and function. WIREs RNA9, e1456 (2018).

    Google Scholar 

  17. Vendramin, R. et al. SAMMSON fosters cancer cell fitness by concertedly enhancing mitochondrial and cytosolic translation. Nat. Struct. Mol. Biol. 25, 1035–1046 (2018).

    CAS  PubMed  Google Scholar 

  18. Grote, P. et al. The tissue-specific lncRNA Fendrr is an essential regulator of heart and body wall development in the mouse. Dev. Cell 24, 206–214 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Huarte, M. et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 142, 409–419 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Andersen, R. E. et al. The long noncoding RNA Pnky is a trans-acting regulator of cortical development in vivo. Dev. Cell 49, 632–642.e7 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Werner, M. S. & Ruthenburg, A. J. Nuclear fractionation reveals thousands of chromatin-tethered noncoding RNAs adjacent to active genes. Cell Rep. 12, 1089–1098 (2015). This study uses nuclear fractionations to show that the majority of lncRNAs, including many previously unannotated intergenic transcripts, are enriched in the chromatin fraction. These chromatin-enriched lncRNAs are dependent on Pol II elongation for their association with chromatin.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Derrien, T. et al. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res. 22, 1775–1789 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Pauli, A. et al. Systematic identification of long noncoding RNAs expressed during zebrafish embryogenesis. Genome Res. 22, 577–591 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Ulitsky, I. Evolution to the rescue: using comparative genomics to understand long non-coding RNAs. Nat. Rev. Genet. 17, 601–614 (2016).

    CAS  PubMed  Google Scholar 

  25. 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 

  26. Lewandowski, J. P. et al. The Firre locus produces a trans-acting RNA molecule that functions in hematopoiesis. Preprint at bioRxiv https://doi.org/10.1101/648279 (2019).

  27. Clemson, C. M. et al. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol. Cell 33, 717–726 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Naganuma, T. et al. Alternative 3′-end processing of long noncoding RNA initiates construction of nuclear paraspeckles. EMBO J. 31, 4020–4034 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Rom, A. et al. Regulation of CHD2 expression by the Chaserr long noncoding RNA is essential for viability. Nat. Commun. 10, 5092 (2019).

    PubMed  PubMed Central  Google Scholar 

  30. Isoda, T. et al. Non-coding transcription instructs chromatin folding and compartmentalization to dictate enhancer–promoter communication and T cell fate. Cell 171, 103–119.e18 (2017). This study characterizes the enhancer-transcribed ThymoD lncRNA, whose transcription promotes the activity of the underlying enhancer through repositioning it away from the nuclear periphery, demonstrating the interdependence between (lncRNA) transcription, nuclear positioning and enhancer activity.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Perry, R. B., Hezroni, H., Goldrich, M. J. & Ulitsky, I. Regulation of neuroregeneration by long noncoding RNAs. Mol. Cell 72, 553–567 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Kotzin, J. J. et al. The long non-coding RNA Morrbid regulates Bim and short-lived myeloid cell lifespan. Nature 537, 239–243 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Engreitz, J. M. et al. Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 539, 452–455 (2016). This study demonstrates that promoters of some lncRNAs, but also of some mRNAs, affect the expression of neighbouring genes, emphasizing that studies of the functions of cis-acting lncRNAs are also applicable to other transcriptional units.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. VučiĆeviĆ, D., Corradin, O., Ntini, E., Scacheri, P. C. & Ørom, U. A. Long ncRNA expression associates with tissue-specific enhancers. Cell Cycle 14, 253–260 (2015).

    PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Marques, A. C. et al. Chromatin signatures at transcriptional start sites separate two equally populated yet distinct classes of intergenic long noncoding RNAs. Genome Biol. 14, R131 (2013). Through analysis of chromatin modifications at regions of transcription initiation, this study notes two distinct classes of intergenic lncRNAs, one with characteristics of enhancers and one with characteristics of promoters.

    PubMed  PubMed Central  Google Scholar 

  37. Ernst, J. & Kellis, M. Chromatin-state discovery and genome annotation with ChromHMM. Nat. Protoc. 12, 2478–2492 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Gil, N. & Ulitsky, I. Production of spliced long noncoding RNAs specifies regions with increased enhancer activity. Cell Syst. 7, 537–547.e3 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Ulitsky, I., Shkumatava, A., Jan, C. H., Sive, H. & Bartel, D. P. Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell 147, 1537–1550 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Wamstad, J. A. et al. Dynamic and coordinated epigenetic regulation of developmental transitions in the cardiac lineage. Cell 151, 206–220 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Ponjavic, J., Oliver, P. L., Lunter, G. & Ponting, C. P. Genomic and transcriptional co-localization of protein-coding and long non-coding RNA pairs in the developing brain. PLOS Genet. 5, e1000617 (2009).

    PubMed  PubMed Central  Google Scholar 

  42. Schwanhausser, B. et al. Global quantification of mammalian gene expression control. Nature 473, 337–342 (2011).

    PubMed  Google Scholar 

  43. Kutter, C. et al. Rapid turnover of long noncoding RNAs and the evolution of gene expression. PLOS Genet. 8, e1002841 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Werner, M. S. et al. Chromatin-enriched lncRNAs can act as cell-type specific activators of proximal gene transcription. Nat. Struct. Mol. Biol. 24, 596–603 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. 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 

  46. Schertzer, M. D. et al. lncRNA-induced spread of polycomb controlled by genome architecture, RNA abundance, and CpG island DNA. Mol. Cell 75, 523–537.e10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Ballarino, M. et al. Deficiency in the nuclear long noncoding RNA causes myogenic defects and heart remodeling in mice. EMBO J. 37, e99697 (2018).

    PubMed  PubMed Central  Google Scholar 

  48. Groff, A. F., Barutcu, A. R., Lewandowski, J. P. & Rinn, J. L. Enhancers in the Peril lincRNA locus regulate distant but not local genes. Genome Biol. 19, 219 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Amaral, P. P. et al. Genomic positional conservation identifies topological anchor point RNAs linked to developmental loci. Genome Biol. 19, 32 (2018).

    PubMed  PubMed Central  Google Scholar 

  50. Tan, J. Y. et al. Cis-acting complex-trait-associated lincRNA expression correlates with modulation of chromosomal architecture. Cell Rep. 18, 2280–2288 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  52. Ma, L., Bajic, V. B. & Zhang, Z. On the classification of long non-coding RNAs. RNA Biol. 10, 925–933 (2013).

    PubMed  Google Scholar 

  53. Fanucchi, S. et al. Immune genes are primed for robust transcription by proximal long noncoding RNAs located in nuclear compartments. Nat. Genet. 51, 138–150 (2019). This article presents a well-studied example of a lncRNA that activates expression of its target gene through recruitment of activating factors — in this case, the WDR5–MLL complex — to the target promoter, utilizing pre-existing spatial proximity.

    CAS  PubMed  Google Scholar 

  54. 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). This work is one of the first examples of an enhancer-transcribed lncRNA that affects the expression of its target gene through promoting the formation of enhancer–promoter chromatin loops, in this case through recruitment of CTCF.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Sleutels, F., Zwart, R. & Barlow, D. P. The non-coding Air RNA is required for silencing autosomal imprinted genes. Nature 415, 810–813 (2002).

    CAS  PubMed  Google Scholar 

  56. Kherdjemil, Y. et al. Evolution of Hoxa11 regulation in vertebrates is linked to the pentadactyl state. Nature 539, 89–92 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. 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 

  58. Tahira, A. C. et al. Long noncoding intronic RNAs are differentially expressed in primary and metastatic pancreatic cancer. Mol. Cancer 10, 141 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Anderson, K. M. et al. Transcription of the non-coding RNA upperhand controls Hand2 expression and heart development. Nature 539, 433–436 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Frank, S. et al. yylncT defines a class of divergently transcribed lncRNAs and safeguards the T-mediated mesodermal commitment of human PSCs. Cell Stem Cell 24, 318–327.e8 (2019).

    CAS  PubMed  Google Scholar 

  61. Martens, J. A., Laprade, L. & Winston, F. Intergenic transcription is required to repress the Saccharomyces cerevisiae SER3 gene. Nature 429, 571–574 (2004).

    CAS  PubMed  Google Scholar 

  62. Bergmann, J. H. et al. Regulation of the ESC transcriptome by nuclear long noncoding RNAs. Genome Res. 25, 1336–1346 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Heintzman, N. D. et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459, 108–112 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Xi, H. et al. Identification and characterization of cell type-specific and ubiquitous chromatin regulatory structures in the human genome. PLOS Genet. 3, e136 (2007).

    PubMed  PubMed Central  Google Scholar 

  65. Chen, H., Du, G., Song, X. & Li, L. Non-coding transcripts from enhancers: new insights into enhancer activity and gene expression regulation. Genomics Proteomics Bioinformatics 15, 201–207 (2017).

    PubMed  PubMed Central  Google Scholar 

  66. Ding, M. et al. Enhancer RNAs (eRNAs): new insights into gene transcription and disease treatment. J. Cancer 9, 2334–2340 (2018).

    PubMed  PubMed Central  Google Scholar 

  67. Li, W., Notani, D. & Rosenfeld, M. G. Enhancers as non-coding RNA transcription units: recent insights and future perspectives. Nat. Rev. Genet. 17, 207–223 (2016).

    CAS  PubMed  Google Scholar 

  68. Koch, F. et al. Transcription initiation platforms and GTF recruitment at tissue-specific enhancers and promoters. Nat. Struct. Mol. Biol. 18, 956–963 (2011).

    CAS  PubMed  Google Scholar 

  69. 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 

  70. Nozawa, R.-S. et al. SAF-A regulates interphase chromosome structure through oligomerization with chromatin-associated RNAs. Cell 169, 1214–1227.e18 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Xiao, R. et al. Nuclear matrix factor hnRNP U/SAF-A exerts a global control of alternative splicing by regulating U2 snRNP maturation. Mol. Cell 45, 656–668 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Elling, R. et al. Genetic models reveal cis and trans immune-regulatory activities for lincRNA-Cox2. Cell Rep. 25, 1511–1524.e6 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Han, X. et al. The lncRNA Hand2os1/Uph locus orchestrates heart development through regulation of precise expression of Hand2. Development 146, dev176198 (2019).

    CAS  Google Scholar 

  74. Ntini, E. et al. Long ncRNA A-ROD activates its target gene DKK1 at its release from chromatin. Nat. Commun. 9, 1636 (2018).

    PubMed  PubMed Central  Google Scholar 

  75. Krawczyk, M. & Emerson, B. M. p50-associated COX-2 extragenic RNA (PACER) activates COX-2 gene expression by occluding repressive NF-κB complexes. eLife 3, e01776 (2014).

    PubMed  PubMed Central  Google Scholar 

  76. Wang, K. C. et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 472, 120–124 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Tan, J. Y., Biasini, A., Young, R. S. & Marques, A. An unexpected contribution of lincRNA splicing to enhancer function. Preprint at bioRxiv https://doi.org/10.1101/287706 (2018).

  78. Haerty, W. & Ponting, C. P. Unexpected selection to retain high GC content and splicing enhancers within exons of multiexonic lncRNA loci. RNA 21, 333–346 (2015).

    PubMed  Google Scholar 

  79. Schüler, A., Ghanbarian, A. T. & Hurst, L. D. Purifying selection on splice-related motifs, not expression level nor RNA folding, explains nearly all constraint on human lincRNAs. Mol. Biol. Evol. 31, 3164–3183 (2014).

    PubMed  PubMed Central  Google Scholar 

  80. Lamond, A. I. & Spector, D. L. Nuclear speckles: a model for nuclear organelles. Nat. Rev. Mol. Cell Biol. 4, 605–612 (2003).

    CAS  PubMed  Google Scholar 

  81. Kim, J., Khanna, N. & Belmont, A. S. Transcription amplification by nuclear speckle association. Preprint at bioRxiv https://doi.org/10.1101/604298 (2019).

  82. Ding, F. & Elowitz, M. B. Constitutive splicing and economies of scale in gene expression. Nat. Struct. Mol. Biol. 26, 424–432 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Fong, Y. W. & Zhou, Q. Stimulatory effect of splicing factors on transcriptional elongation. Nature 414, 929–933 (2001).

    CAS  PubMed  Google Scholar 

  84. Sahakyan, A., Yang, Y. & Plath, K. The role of xist in X-chromosome dosage compensation. Trends Cell Biol. 28, 999–1013 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Robert Finestra, T. & Gribnau, J. X chromosome inactivation: silencing, topology and reactivation. Curr. Opin. Cell Biol. 46, 54–61 (2017).

    CAS  PubMed  Google Scholar 

  86. da Rocha, S. T. & Heard, E. Novel players in X inactivation: insights into Xist-mediated gene silencing and chromosome conformation. Nat. Struct. Mol. Biol. 24, 197–204 (2017).

    PubMed  Google Scholar 

  87. Swiezewski, S., Liu, F., Magusin, A. & Dean, C. Cold-induced silencing by long antisense transcripts of an arabidopsis polycomb target. Nature 462, 799–802 (2009).

    CAS  PubMed  Google Scholar 

  88. Rosa, S., Duncan, S. & Dean, C. Mutually exclusive sense–antisense transcription at FLC facilitates environmentally induced gene repression. Nat. Commun. 7, 13031 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Whittaker, C. & Dean, C. The FLC locus: a platform for discoveries in epigenetics and adaptation. Annu. Rev. Cell Dev. Biol. 33, 555–575 (2017).

    CAS  PubMed  Google Scholar 

  90. Beckedorff, F. C. et al. The intronic long noncoding RNA ANRASSF1 recruits PRC2 to the RASSF1A promoter, reducing the expression of RASSF1A and increasing cell proliferation. PLOS Genet. 9, e1003705 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Cho, S. W. et al. Promoter of lncRNA gene PVT1 is a tumor-suppressor DNA boundary element. Cell 173, 1398–1412.e22 (2018). This article presents an example of a lncRNA gene that represses its target through enhancer competition: the PVT1 promoter competes with the MYC promoter on association with and activation by proximally situated enhancers.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Wang, F. et al. Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizing NOP2. Hepatology 60, 1278–1290 (2014).

    CAS  PubMed  Google Scholar 

  93. Tseng, Y. Y. et al. PVT1 dependence in cancer with MYC copy-number increase. Nature 512, 82–86 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Xiao, M., Feng, Y., Liu, C. & Zhang, Z. Prognostic values of long noncoding RNA PVT1 in various carcinomas: an updated systematic review and meta-analysis. Cell Prolif. 51, e12519 (2018).

    PubMed  PubMed Central  Google Scholar 

  95. Latos, P. A. et al. Airn transcriptional overlap, but not its lncRNA products, induces imprinted Igf2r silencing. Science 338, 1469–1472 (2012). This study is a compelling demonstration that transcription through specific regions of a lncRNA gene can be functionally significant, and thus demonstrates the importance of careful selection of integration sites for sequences that interfere with transcription.

    CAS  PubMed  Google Scholar 

  96. Hongay, C. F., Grisafi, P. L., Galitski, T. & Fink, G. R. Antisense transcription controls cell fate in Saccharomyces cerevisiae. Cell 127, 735–745 (2006).

    CAS  PubMed  Google Scholar 

  97. van Werven, F. J. et al. Transcription of two long noncoding RNAs mediates mating-type control of gametogenesis in budding yeast. Cell 150, 1170–1181 (2012).

    PubMed  PubMed Central  Google Scholar 

  98. Bumgarner, S. L., Dowell, R. D., Grisafi, P., Gifford, D. K. & Fink, G. R. Toggle involving cis-interfering noncoding RNAs controls variegated gene expression in yeast. Proc. Natl Acad. Sci. USA 106, 18321–18326 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Thebault, P. et al. Transcription regulation by the noncoding RNA SRG1 requires Spt2-dependent chromatin deposition in the wake of RNA polymerase II. Mol. Cell. Biol. 31, 1288–1300 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Canzio, D. et al. Antisense lncRNA transcription mediates DNA demethylation to drive stochastic protocadherin α promoter choice. Cell 177, 639–653.e15 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Bumgarner, S. L. et al. Single-cell analysis reveals that noncoding RNAs contribute to clonal heterogeneity by modulating transcription factor recruitment. Mol. Cell 45, 470–482 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Ritter, N. et al. The lncRNA locus Handsdown regulates cardiac gene programs and is essential for early mouse development. Dev. Cell 50, 644–657.e8 (2019).

    CAS  PubMed  Google Scholar 

  103. Richard, J. L. C. & Ogawa, Y. Understanding the complex circuitry of lncRNAs at the X-inactivation center and its implications in disease conditions. Curr. Top. Microbiol. Immunol. 394, 1–27 (2016).

    PubMed  Google Scholar 

  104. Tajul-Arifin, K. et al. Identification and analysis of chromodomain-containing proteins encoded in the mouse transcriptome. Genome Res. 13, 1416–1429 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Paralkar, V. R. et al. Unlinking an lncRNA from its associated cis element. Mol. Cell 62, 104–110 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Alexanian, M. et al. A transcribed enhancer dictates mesendoderm specification in pluripotency. Nat. Commun. 8, 1806 (2017).

    PubMed  PubMed Central  Google Scholar 

  107. Carmona, S., Lin, B., Chou, T., Arroyo, K. & Sun, S. lncRNA Jpx induces xist expression in mice using both trans and cis mechanisms. PLOS Genet. 14, e1007378 (2018).

    PubMed  PubMed Central  Google Scholar 

  108. Nagano, T. et al. The Air noncoding RNA epigenetically silences transcription by targeting G9a to chromatin. Science 322, 1717–1720 (2008).

    CAS  PubMed  Google Scholar 

  109. Sigova, A. A. et al. Transcription factor trapping by RNA in gene regulatory elements. Science 350, 978–981 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Khalil, A. M. et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc. Natl Acad. Sci. USA 106, 11667–11672 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. 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 

  112. 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). This study finds that PRC2 binds RNA with high affinity but low specificity both in vitro and in vivo, with its promiscuous binding patterns correlating with RNA lengths or expression levels, and with no apparent specificity for biologically significant interactions. This casts doubt on the proposed role of many lncRNAs in recruiting PRC2 to their transcription sites.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Cifuentes-Rojas, C., Hernandez, A. J., Sarma, K. & Lee, J. T. Regulatory interactions between RNA and polycomb repressive complex 2. Mol. Cell 55, 171–185 (2014). This study notes that whereas the EZH2 subunit of PRC2 displays high-affinity RNA binding but with low specificity, other subunits – mostly EED – increase the selectivity of the complex’s RNA interactions.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Wang, X. et al. Targeting of polycomb repressive complex 2 to RNA by short repeats of consecutive guanines. Mol. Cell 65, 1056–1067.e5 (2017).

    CAS  PubMed  Google Scholar 

  115. Colognori, D., Sunwoo, H., Kriz, A. J., Wang, C.-Y. & Lee, J. T. Xist deletional analysis reveals an interdependency between Xist RNA and polycomb complexes for spreading along the inactive X. Mol. Cell 74, 101–117.e10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Baltz, A. G. et al. The mRNA-bound proteome and its global occupancy profile on protein-coding transcripts. Mol. Cell 46, 674–690 (2012).

    CAS  PubMed  Google Scholar 

  117. Perez-Perri, J. I. et al. Discovery of RNA-binding proteins and characterization of their dynamic responses by enhanced RNA interactome capture. Nat. Commun. 9, 4408 (2018).

    PubMed  PubMed Central  Google Scholar 

  118. He, C. et al. High-resolution mapping of RNA-binding regions in the nuclear proteome of embryonic stem cells. Mol. Cell 64, 416–430 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Castello, A. et al. Comprehensive identification of RNA-binding domains in human cells. Mol. Cell 63, 696–710 (2016). In this study, the authors develop RBDmap, a strategy for identifying RNA-binding regions in proteins through ultraviolet crosslinking, oligo(dT) capture and identification of crosslinked peptides using mass spectrometry. This and similar methods detailed in Baltz et al., Perez-Perri et al and He et al. unravelled a myriad of known and novel RNA-binding proteins and domains.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Hansen, A. S. et al. Distinct classes of chromatin loops revealed by deletion of an RNA-binding region in CTCF. Mol. Cell 76, 395–411.e13 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Sun, S. et al. Jpx RNA activates Xist by evicting CTCF. Cell 153, 1537–1551 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Kaneko, S., Son, J., Bonasio, R., Shen, S. S. & Reinberg, D. Nascent RNA interaction keeps PRC2 activity poised and in check. Genes Dev. 28, 1983–1988 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Beltran, M. et al. The interaction of PRC2 with RNA or chromatin is mutually antagonistic. Genome Res. 26, 896–907 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Hosogane, M., Funayama, R., Shirota, M. & Nakayama, K. Lack of transcription triggers H3K27me3 accumulation in the gene body. Cell Rep. 16, 696–706 (2016).

    CAS  PubMed  Google Scholar 

  125. Beltran, M. et al. G-tract RNA removes polycomb repressive complex 2 from genes. Nat. Struct. Mol. Biol. 26, 899–900 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Sims, R. J. Elongation by RNA polymerase II: the short and long of it. Genes Dev. 18, 2437–2468 (2004).

    CAS  PubMed  Google Scholar 

  127. Brandão, H. B. et al. RNA polymerases as moving barriers to condensin loop extrusion. Proc. Natl Acad. Sci. USA 116, 20489–20499 (2019).

    PubMed  PubMed Central  Google Scholar 

  128. Hsieh, T.-H. S. et al. Resolving the 3D landscape of transcription-linked mammalian chromatin folding. Preprint at bioRxiv https://doi.org/10.1101/638775 (2019).

  129. Heinz, S. et al. Transcription elongation can affect genome 3D structure. Cell 174, 1522–1536.e22 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Gu, B. et al. Transcription-coupled changes in nuclear mobility of mammalian cis-regulatory elements. Science 359, 1050–1055 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Nagashima, R. et al. Single nucleosome imaging reveals loose genome chromatin networks via active RNA polymerase II. J. Cell Biol. 218, 1511–1530 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Kopp, F. & Mendell, J. T. Functional classification and experimental dissection of long noncoding RNAs. Cell 172, 393–407 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Sander, J. D. & Joung, J. K. CRISPR–Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 32, 347–355 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Dominguez, A. A., Lim, W. A. & Qi, L. S. Beyond editing: repurposing CRISPR–Cas9 for precision genome regulation and interrogation. Nat. Rev. Mol. Cell Biol. 17, 5–15 (2016).

    CAS  PubMed  Google Scholar 

  135. Lavalou, P. et al. Strategies for genetic inactivation of long noncoding RNAs in zebrafish. RNA 25, 897–904 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Fulco, C. P. et al. Systematic mapping of functional enhancer–promoter connections with CRISPR interference. Science 354, 769–773 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Simeonov, D. R. et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature 549, 111–115 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Abudayyeh, O. O. et al. RNA targeting with CRISPR–Cas13. Nature 550, 280–284 (2017).

    PubMed  PubMed Central  Google Scholar 

  139. Cox, D. B. T. et al. RNA editing with CRISPR–Cas13. Science 358, 1019–1027 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Shechner, D. M., Hacisuleyman, E., Younger, S. T. & Rinn, J. L. Multiplexable, locus-specific targeting of long RNAs with CRISPR–Display. Nat. Methods 12, 664–670 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Carullo, N. V. N. et al. Enhancer RNAs are necessary and sufficient for activity-dependent neuronal gene transcription. Preprint at bioRxiv https://doi.org/10.1101/270967 (2018).

  142. Tuck, A. C. et al. Distinctive features of lincRNA gene expression suggest widespread RNA-independent functions. Life Sci. Alliance 1, e201800124 (2018).

    PubMed  PubMed Central  Google Scholar 

  143. Nomura, Y., Zhou, L., Miu, A. & Yokobayashi, Y. Controlling mammalian gene expression by allosteric hepatitis delta virus ribozymes. ACS Synth. Biol. 2, 684–689 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Nojima, T. et al. Mammalian NET-seq reveals genome-wide nascent transcription coupled to RNA processing. Cell 161, 526–540 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Sarshad, A. A. et al. Argonaute–miRNA complexes silence target mRNAs in the nucleus of mammalian stem cells. Mol. Cell 71, 1040–1050.e8 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Wee, K. B. et al. Dynamics of co-transcriptional pre-mRNA folding influences the induction of dystrophin exon skipping by antisense oligonucleotides. PLOS ONE 3, e1844 (2008).

    PubMed  PubMed Central  Google Scholar 

  147. Sioud, M. Promises and challenges in developing RNAi as a research tool and therapy. Methods Mol. Biol. 703, 173–187 (2011).

    CAS  PubMed  Google Scholar 

  148. Stojic, L. et al. Specificity of RNAi, LNA and CRISPRi as loss-of-function methods in transcriptional analysis. Nucleic Acids Res. 46, 5950–5966 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Engreitz, J., Lander, E. S. & Guttman, M. RNA antisense purification (RAP) for mapping RNA interactions with chromatin. Methods Mol. Biol. 1262, 183–197 (2015).

    CAS  PubMed  Google Scholar 

  150. Li, X. & Fu, X.-D. Chromatin-associated RNAs as facilitators of functional genomic interactions. Nat. Rev. Genet. 20, 503–519 (2019). This work is an excellent recent review, particularly useful for those looking for detailed information about techniques used to identify chromatin binding sites of lncRNAs.

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Li, X. et al. GRID-seq reveals the global RNA–chromatin interactome. Nat. Biotechnol. 35, 940–950 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Bell, J. C. et al. Chromatin-associated RNA sequencing (ChAR-seq) maps genome-wide RNA-to-DNA contacts. eLife 7, e27024 (2018).

    PubMed  PubMed Central  Google Scholar 

  153. Sridhar, B. et al. Systematic mapping of RNA–chromatin interactions in vivo. Curr. Biol. 27, 610–612 (2017).

    CAS  PubMed  Google Scholar 

  154. Bonetti, A. et al. RADICL-seq identifies general and cell type-specific principles of genome-wide RNA–chromatin interactions. Preprint at bioRxiv https://doi.org/10.1101/681924 (2019).

  155. Shao, W. et al. U1 snRNP regulates chromatin retention of noncoding RNAs. Preprint at bioRxiv https://doi.org/10.1101/310433 (2018).

  156. Lubelsky, Y. & Ulitsky, I. Sequences enriched in Alu repeats drive nuclear localization of long RNAs in human cells. Nature 555, 107–111 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Shukla, C. J. et al. High-throughput identification of RNA nuclear enrichment sequences. EMBO J. 37, e9845 (2018).

    Google Scholar 

  158. Matsui, M. & Corey, D. R. Non-coding RNAs as drug targets. Nat. Rev. Drug Discov. 16, 167–179 (2017).

    CAS  PubMed  Google Scholar 

  159. Dao, L. T. M. et al. Genome-wide characterization of mammalian promoters with distal enhancer functions. Nat. Genet. 49, 1073–1081 (2017).

    CAS  PubMed  Google Scholar 

  160. Carelli, F. N., Liechti, A., Halbert, J., Warnefors, M. & Kaessmann, H. Repurposing of promoters and enhancers during mammalian evolution. Nat. Commun. 9, 4066 (2018). This study identifies prevalent repurposing of enhancers to promoters (or vice versa) during evolution, demonstrating the large potential that enhancers serve in creating novel transcriptional units, including lncRNAs.

    PubMed  PubMed Central  Google Scholar 

  161. Marques, A. C. et al. Evidence for conserved post-transcriptional roles of unitary pseudogenes and for frequent bifunctionality of mRNAs. Genome Biol. 13, R102 (2012).

    PubMed  PubMed Central  Google Scholar 

  162. Hezroni, H. et al. A subset of conserved mammalian long non-coding RNAs are fossils of ancestral protein-coding genes. Genome Biol. 18, 162 (2017).

    PubMed  PubMed Central  Google Scholar 

  163. Cabili, M. N. et al. Localization and abundance analysis of human lncRNAs at single-cell and single-molecule resolution. Genome Biol. 16, 20 (2015).

    PubMed  PubMed Central  Google Scholar 

  164. Carlevaro-Fita, J. & Johnson, R. Global positioning system: understanding long noncoding RNAs through subcellular localization. Mol. Cell 73, 869–883 (2019).

    CAS  PubMed  Google Scholar 

  165. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Roadmap Epigenomics Consortium. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    PubMed Central  Google Scholar 

  167. 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 

  168. Minajigi, A. et al. Chromosomes. A comprehensive Xist interactome reveals cohesin repulsion and an RNA-directed chromosome conformation. Science https://doi.org/10.1126/science.aab2276 (2015).

    Google Scholar 

  169. Sarma, K. et al. ATRX directs binding of PRC2 to Xist RNA and polycomb targets. Cell 159, 869–883 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Chu, C. et al. Systematic discovery of Xist RNA binding proteins. Cell 161, 404–416 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. McHugh, C. A. et al. The Xist lncRNA interacts directly with SHARP to silence transcription through HDAC3. Nature 521, 232–236 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Hasegawa, Y. et al. The matrix protein hnRNP U is required for chromosomal localization of Xist RNA. Dev. Cell 19, 469–476 (2010).

    CAS  PubMed  Google Scholar 

  173. Dinger, M. E. et al. Long noncoding RNAs in mouse embryonic stem cell pluripotency and differentiation. Genome Res. 18, 1433–1445 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Luo, S. et al. Divergent lncRNAs regulate gene expression and lineage differentiation in pluripotent cells. Cell Stem Cell 18, 637–652 (2016).

    CAS  PubMed  Google Scholar 

  175. Knauss, J. L. et al. Long noncoding RNA Sox2ot and transcription factor YY1 co-regulate the differentiation of cortical neural progenitors by repressing Sox2. Cell Death Dis. 9, 799 (2018).

    PubMed  PubMed Central  Google Scholar 

  176. Jeon, Y. & Lee, J. T. YY1 tethers Xist RNA to the inactive X nucleation center. Cell 146, 119–133 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank A. Shkumatava, S. Nakagawa, L. Chen, C. Ross, H. Hezroni, M. Goldrich and members of the Ulitsky laboratory for helpful discussions and comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Igor Ulitsky.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Genetics thanks C. Ponting and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

Enhancers

DNA regulatory elements that activate gene transcription. Enhancers can operate from nearby or within their targets, or across large genomic distances.

Transcription factors

(TFs). Proteins that bind to specific DNA sequence motifs found within regulatory DNA elements — either promoters or enhancers — to modulate gene expression.

Chromatin remodellers

Proteins that regulate gene expression by altering chromatin structure. Two main groups exist: those that mediate post-translational modifications of histones; and ATP-dependent chromatin remodellers, which regulate the association with and location of nucleosomes along the DNA.

Super-enhancer

A particularly active and cell type-specific enhancer. Demarcated by high levels of chromatin modifications such as histone 3 lysine 27 acetylation (H3K27ac) and long sequence stretches bound by transcription factors and coactivators such as Mediator.

Topologically associating domain

(TAD). A genomic region with an average size of ~1 Mb characterized by high-density chromatin interactions. Sequences within TADs tend to form interactions with one another but less so with sequences in other TADs.

Enhancer RNAs

(eRNAs). A species of bidirectional, unstable non-coding RNAs produced at enhancers. Considered a hallmark of active enhancers and sometimes used for enhancer annotation.

CTCF

A transcription factor that acts primarily in chromatin 3D architecture regulation, through anchoring long-range chromatin loops and demarcating topologically associating domain boundaries.

Polycomb repressive complex 2

(PRC2). A histone methyltransferase protein complex that induces trimethylation of histone 3 lysine 27 (H3K27), a histone modification associated with long-term epigenetic silencing.

Vernalization

The process of induction of plant flowering, brought on by exposure to prolonged cold temperatures.

Enhancer competition

Two (or more) transcriptional units that can be activated by the same enhancer, and which compete over direct binding to and activation by that enhancer.

Transcriptional interference

A process whereby transcription through one genomic region interferes with transcription of a nearby (often overlapping) locus, for example, by curbing the recruitment of trans factors such as transcription factors or chromatin remodellers, or through deposition of chromatin modifications incompatible with transcription initiation.

Auto-regulatory feedback loops

A type of transcriptional regulation network in which a gene product regulates its own levels, for example, a transcription factor which binds its own locus and activates (or represses) transcription.

Disordered regions

Proteins or regions within proteins that do not adopt an ordered or well-defined 3D structure. These regions can serve as linkers between structured regions, or be functional themselves.

CRISPR–Cas9

A bacterial immune mechanism whereby a Cas9 protein uses short guide RNA (gRNA) sequences to target and cleave foreign DNA. CRISPR–Cas9 can be used for gene editing, by ectopic expression of both Cas9 and a gRNA that targets the gene of interest.

CRISPR display

Utilization of CRISPR–Cas9 for the recruitment of non-protein components. For example, long non-coding RNA sequences can be fused to the guide RNA and be brought to the target locus via ‘dead’ Cas9.

Self-cleaving ribozyme sequences

RNA sequences that can catalyse a reaction that would cut their own RNA.

RNA interference

(RNAi). Short non-coding RNA molecules — either microRNAs or short interfering RNAs — bind to complementary sequences in the target genes, leading to translation inhibition or target RNA degradation.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gil, N., Ulitsky, I. Regulation of gene expression by cis-acting long non-coding RNAs. Nat Rev Genet 21, 102–117 (2020). https://doi.org/10.1038/s41576-019-0184-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-019-0184-5

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