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:

Chromatin-enriched lncRNAs can act as cell-type specific activators of proximal gene transcription

Abstract

We recently described a new class of long noncoding RNAs (lncRNAs) that are distinguished by especially tight chromatin association and whose presence is strongly correlated to expression of nearby genes. Here, we examine the cis-enhancer mechanism of this class of chromatin-enriched RNA (cheRNA) across multiple human cell lines. cheRNAs are largely cell type specific and provide the most reliable chromatin signature to predict cis-gene transcription in every human cell type examined. Targeted depletion of three cheRNAs decreases expression of their neighboring genes, indicating potential co-activator function, and single-molecule fluorescence in situ hybridization (smFISH) of one cheRNA-distal target gene pair suggests a spatial overlap consistent with a role in chromosome looping. Additionally, the cheRNA HIDALGO stimulates the fetal hemoglobin subunit gamma 1 (HBG1) gene during erythroid differentiation by promoting contacts to a downstream enhancer. Our results suggest that multiple cheRNAs activate proximal lineage-specific gene transcription.

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

Access options

Buy this article

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

Figure 1: Tissue specificity and cis-gene activity of cheRNAs.
Figure 2: cheRNA PVT1 bridges the site of its production to its target gene, MYC.
Figure 3: Examination of cis-enhancer activity of cheRNA–gene pairs.
Figure 4: HIDALGO exhibits hallmarks of a promoter and is induced with HBG1 during erythrogenesis.
Figure 5: HIDALGO promotes HBG1 expression.
Figure 6: HIDALGO couples an enhancer at its TSS to the HBG1 promoter.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Yang, L. et al. lncRNA-dependent mechanisms of androgen-receptor-regulated gene activation programs. Nature 500, 598–602 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Trimarchi, T. et al. Genome-wide mapping and characterization of Notch-regulated long noncoding RNAs in acute leukemia. Cell 158, 593–606 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Feng, J. et al. The Evf-2 noncoding RNA is transcribed from the Dlx-5/6 ultraconserved region and functions as a Dlx-2 transcriptional coactivator. Genes Dev. 20, 1470–1484 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Li, W. et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498, 516–520 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lai, F., Gardini, A., Zhang, A. & Shiekhattar, R. Integrator mediates the biogenesis of enhancer RNAs. Nature 525, 399–403 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Davidovich, C. et al. Toward a consensus on the binding specificity and promiscuity of PRC2 for RNA. Mol. Cell 57, 552–558 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mao, Y.S., Sunwoo, H., Zhang, B. & Spector, D.L. Direct visualization of the co-transcriptional assembly of a nuclear body by noncoding RNAs. Nat. Cell Biol. 13, 95–101 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Engreitz, J.M. et al. Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 539, 452–455 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Petruk, S. et al. Transcription of bxd noncoding RNAs promoted by trithorax represses Ubx in cis by transcriptional interference. Cell 127, 1209–1221 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Schmitt, S., Prestel, M. & Paro, R. Intergenic transcription through a polycomb group response element counteracts silencing. Genes Dev. 19, 697–708 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Melé, M. & Rinn, J.L. “Cat's cradling” the 3D genome by the act of lncRNA transcription. Mol. Cell 62, 657–664 (2016).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bhatt, D.M. et al. Transcript dynamics of proinflammatory genes revealed by sequence analysis of subcellular RNA fractions. Cell 150, 279–290 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wuarin, J. & Schibler, U. Physical isolation of nascent RNA chains transcribed by RNA polymerase II: evidence for cotranscriptional splicing. Mol. Cell. Biol. 14, 7219–7225 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  22. Postepska-Igielska, A. et al. LncRNA Khps1 regulates expression of the proto-oncogene SPHK1 via triplex-mediated changes in chromatin structure. Mol. Cell 60, 626–636 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Sunwoo, H., Wu, J.Y. & Lee, J.T. The Xist RNA-PRC2 complex at 20-nm resolution reveals a low Xist stoichiometry and suggests a hit-and-run mechanism in mouse cells. Proc. Natl. Acad. Sci. USA 112, E4216–E4225 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  25. Ørom, U.A. et al. Long noncoding RNAs with enhancer-like function in human cells. Cell 143, 46–58 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Ernst, J. et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Andersson, R. et al. An atlas of active enhancers across human cell types and tissues. Nature 507, 455–461 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Vilborg, A., Passarelli, M.C., Yario, T.A., Tycowski, K.T. & Steitz, J.A. Widespread inducible transcription downstream of human genes. Mol. Cell 59, 449–461 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Na, J., Furue, M.K. & Andrews, P.W. Inhibition of ERK1/2 prevents neural and mesendodermal differentiation and promotes human embryonic stem cell self-renewal. Stem Cell Res. 5, 157–169 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. de Groot, R.P., Raaijmakers, J.A., Lammers, J.W., Jove, R. & Koenderman, L. STAT5 activation by BCR-Abl contributes to transformation of K562 leukemia cells. Blood 94, 1108–1112 (1999).

    CAS  PubMed  Google Scholar 

  33. Rao, S.S.P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Robinson, P.J. & Rhodes, D. Structure of the '30 nm' chromatin fibre: a key role for the linker histone. Curr. Opin. Struct. Biol. 16, 336–343 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Li, G. et al. Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation. Cell 148, 84–98 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gilbert, L.A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Addya, S. et al. Erythroid-induced commitment of K562 cells results in clusters of differentially expressed genes enriched for specific transcription regulatory elements. Physiol. Genomics 19, 117–130 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Bodine, D.M. & Ley, T.J. An enhancer element lies 3′ to the human A gamma globin gene. EMBO J. 6, 2997–3004 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Purucker, M., Bodine, D., Lin, H., McDonagh, K. & Nienhuis, A.W. Structure and function of the enhancer 3′ to the human A γ globin gene. Nucleic Acids Res. 18, 7407–7415 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Maamar, H., Cabili, M.N., Rinn, J. & Raj, A. linc-HOXA1 is a noncoding RNA that represses Hoxa1 transcription in cis. Genes Dev. 27, 1260–1271 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Dekker, J., Rippe, K., Dekker, M. & Kleckner, N. Capturing chromosome conformation. Science 295, 1306–1311 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Schultz, D.C., Ayyanathan, K., Negorev, D., Maul, G.G. & Rauscher, F.J. III. SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev. 16, 919–932 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Grzybowski, A.T., Chen, Z. & Ruthenburg, A.J. Calibrating ChIP-Seq with nucleosomal internal standards to measure histone modification density genome wide. Mol. Cell 58, 886–899 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hansen, K.H. et al. A model for transmission of the H3K27me3 epigenetic mark. Nat. Cell Biol. 10, 1291–1300 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Andersson, R., Sandelin, A. & Danko, C.G. A unified architecture of transcriptional regulatory elements. Trends Genet. 31, 426–433 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Arner, E. et al. Transcribed enhancers lead waves of coordinated transcription in transitioning mammalian cells. Science 347, 1010–1014 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. O'Sullivan, J.M. et al. Gene loops juxtapose promoters and terminators in yeast. Nat. Genet. 36, 1014–1018 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Guttman, M. et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 477, 295–300 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  54. Penny, G.D., Kay, G.F., Sheardown, S.A., Rastan, S. & Brockdorff, N. Requirement for Xist in X chromosome inactivation. Nature 379, 131–137 (1996).

    Article  CAS  PubMed  Google Scholar 

  55. Kapusta, A. et al. Transposable elements are major contributors to the origin, diversification, and regulation of vertebrate long noncoding RNAs. PLoS Genet. 9, e1003470 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lynch, V.J. et al. Ancient transposable elements transformed the uterine regulatory landscape and transcriptome during the evolution of mammalian pregnancy. Cell Rep. 10, 551–561 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Johnson, R.M. et al. Fetal globin expression in New World monkeys. J. Biol. Chem. 271, 14684–14691 (1996).

    Article  CAS  PubMed  Google Scholar 

  58. Chuong, E.B., Elde, N.C. & Feschotte, C. Regulatory evolution of innate immunity through co-option of endogenous retroviruses. Science 351, 1083–1087 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Harrow, J. et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 22, 1760–1774 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Forrest, A.R. et al. A promoter-level mammalian expression atlas. Nature 507, 462–470 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. Ichetovkin, I.E., Abramochkin, G. & Shrader, T.E. Substrate recognition by the leucyl/phenylalanyl-tRNA-protein transferase. Conservation within the enzyme family and localization to the trypsin-resistant domain. J. Biol. Chem. 272, 33009–33014 (1997).

    Article  CAS  PubMed  Google Scholar 

  62. Vandesompele, J. et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 3 research0034.1–research0034.11 (2002).

  63. Lenhard, B., Sandelin, A. & Carninci, P. Metazoan promoters: emerging characteristics and insights into transcriptional regulation. Nat. Rev. Genet. 13, 233–245 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Siebert, M. & Söding, J. Universality of core promoter elements? Nature 511, E11–E12 (2014).

    Article  CAS  PubMed  Google Scholar 

  65. Guenther, M.G., Levine, S.S., Boyer, L.A., Jaenisch, R. & Young, R.A. A chromatin landmark and transcription initiation at most promoters in human cells. Cell 130, 77–88 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Gilbert, L.A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Chen, B. et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 155, 1479–1491 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hagège, H. et al. Quantitative analysis of chromosome conformation capture assays (3C-qPCR). Nat. Protoc. 2, 1722–1733 (2007).

    Article  PubMed  Google Scholar 

  70. Cordelieres, F.P. & Bolte, S. JACoP v2.0: improving the user experience with co-localization studies. in Proceedings of the 2nd ImageJ User and Developer Conference 174–181 (2008).

  71. Nadadur, R.D. et al. Pitx2 modulates a Tbx5-dependent gene regulatory network to maintain atrial rhythm. Science Translational Medicine 8, 354ra115 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Hattori, T. et al. Antigen clasping by two antigen-binding sites of an exceptionally specific antibody for histone methylation. Proc. Natl. Acad. Sci. USA 113, 2092–2097 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Rohland, N. & Reich, D. Cost-effective, high-throughput DNA sequencing libraries for multiplexed target capture. Genome Res. 22, 939–946 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful for ChIP-seq data from ENCODE, in particular that deposited by the Snyder and Bernstein labs. We thank the following University of Chicago Core Facility personnel: P. Faber and H. Whitehurst in Functional Genomics for Illumina sequencing and D. Leclerc in Flow Cytometry for FACs training. K562 cells with dCas9-KRAB stably integrated into the genome were generously provided by L. Gilbert and J. Weissman. We would also like to thank V. Lynch for helpful discussions regarding cheRNAs and transposable elements. These studies were supported by Stanley Manne Children's Research Institute's Grant to V.G. We thank Y. Galat for technical help in ESC culture. M.S.W. is supported by the Chicago Biotechnology Consortium with support from The Searle Funds at The Chicago Community Trust; A.J.R. is supported by the Ellison Medical Foundation (AG-NS-1118-13), NIH (R01-GM115945), and American Cancer Society (130230-RSG-16-248-01-DMC).

Author information

Authors and Affiliations

Authors

Contributions

A.J.R. and M.S.W. designed the study and wrote the paper with valuable input from all of the other authors. M.S.W. performed the experiments for Figures 1 and 2 and the initial experiments for Figures 3, 4, and 5. M.A.S. repeated most of the Figures 3, 4, and 5 experiments in higher replicate with assistance from M.S.W., and performed 3C with ASO-treated cells. R.N.S. and A.T.G. performed ICeChIP experiments. R.D.N. performed the luciferase assay to examine enhancer activity of the HIDALGO TSS with oversight from I.P.M. V.G. cultured H1 ESCs used.

Corresponding author

Correspondence to Alexander J Ruthenburg.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 In vitro transcribed RNA standards establish a calibration curve for quantitation of nuclear RNAs.

a, Agarose gel of spike-in standards demonstrates purity of transcripts. b, Scatter plot of spike-in standards demonstrates a linear relationship with molecules added and abundance measured by RT-qPCR. c, Scatter plot of RNA-seq reads (y-axis) versus molecules spiked-in per cell (x-axis) generally recapitulates a linear relationship in all libraries, the slopes of these curves were used to compute molecules/cell of each cheRNA. As a comparison the highest read value in each library was back calculated by the fold-dilution to a theoretical read value (green lines).

Supplementary Figure 2 Properties of cheRNAs in K562 and H1 ESCs.

a, A tabular summary of cheRNA properties as compared to lncRNAs59 and eRNAs29 by cell type (see methods). Percent overlaps are calculated as overlap of features in columns (‘-a’ in bedtools intersect) to features in rows (‘-b’). Cell-type specific lncRNAs and eRNAs were determined if they exhibited coverage from the respective cell types in our RNA-seq data. b, Coding Potential Assessment Tool (CPAT) of H1 and K562 cheRNAs compared to all Gencode (v23) lincRNAs and mRNAs (not defined by cell-type). c, Same as in Fig. 1e but with more comparisons, including categorization by location of cheRNAs relative to neighboring genes, and TADs containing cheRNAs, and the expression of all genes within TADs. Gene-gene pair analysis in TADs is representative of nearest gene expression of randomly chosen 3,293 genes (same as number of cheRNAs) within TADs. P values calculated relative to all cheRNAs, ****P<1x10-16, ***P<1x10-5, **P<0.001, *P<0.05, by Mann-Whitney U test. d, Fraction of our cheRNA set that overlap with CAGE peaks from existing data from FANTOM81. e, Gene ontology (GO) performed with GREAT on the closest neighboring gene for each cheRNA suggests lineage-specific functions for H1 and K562 cheRNAs. The categories with significant hypergeometric enrichment relative to the whole genome background are shown for each cell line measured with raw P values and annotation categories comprising 50-150 genes. See Supplementary Bioinformatics Note for details.

Supplementary Figure 3 Assessment of specificity in CRISPRi experiments and Hi-C data in K562 cells demonstrate cheRNA-gene pairs lie within the boundaries of topological domains.

a, RT-qPCR of GAPDH from CRISPRi experiments in Figure 2d-f demonstrates unchanged or inconsistent changes in GAPDH, in contrast to nearby genes. b, CRISPRi in HEK293 cells at cheRNAs targets BONIFACIO (b) and PAINE c, that are not expressed does not affect neighboring gene expression (n=3 biological replicates). IL6-ILYCH gene-cheRNA pair could not be queried because there is no detectable IL6 in HEK293 cells. d-f, Three-dimensional chromosome contacts measured by Hi-C in K562 cells from Rao et al., indicated by red color-scale33. X and y axes are the same chromosome span (indicated), and off diagonal contact enrichment represents a contact domain. CheRNA-gene pairs from figure are indicated with dotted lines.

Supplementary Figure 4 Specificity of cheRNAs through differentiation and characterization of HIDALGO transcription.

a, Overlap of cheRNAs in K562 cells in un-induced and hemin-induced states demonstrate a majority overlap, in contrast to between cell lines in general (Fig. 1d). b, RNA-seq of chromatin pellet (CPE) (purple shades) and soluble nuclear extract (SNE) (green shades) from K562 cells in +/- hemin conditions, and H1 hESC overlaid at the HBG1-HIDALGO locus. c, Schematic of HIDALGO 5’/3’ RACE products with indicated genetic features (PAS = polyadenylation sequence, TATA = TATA box, TFBS = transcription factor binding site). Red stripes indicate in-frame stop codons. Primer sets for the deconvolution of relative amounts of each isoform indicated in cyan on the gene structure are available in Supplementary Table 2. Note that the TATA just upstream of HIDALGO transcript #4 is “TATAAG”. d, 3’ RACE of HBG1. Inset represents densitometry of HBG1 3’ RACE run on an agarose gel stained with ethidium bromide. e, 5’ RACE of HBG1. Agarose gel stained with ethidium bromide depicts different HIDALGO transcripts during a hemin time-course. 3′ and 5′ RACE were performed with SMARTER 5’/3’ RACE kit (Clontech). f, RT-qPCR of HIDALGO with a panel of qPCR primers designed to assess individual transcript isoforms before and after two hours of hemin induction. Although isoform #4 cannot be uniquely probed, its several-fold induction can be inferred from it comprising little of the resting HIDALGO pool, but accounting for ~1/3 of the #1-4 increase upon induction. (n = 3, error presented is S.E.M.). g, Melting temperatures for amplicons derived from qPCR primers (Supplementary Table 2) designed to differentiate between HBG1 and HBG2 transcription by amplifying unique regions within each intron 2 demonstrate specificity.

Supplementary Figure 5 Examination of the interconnection of HIDALGO and HBG1/2 RNA products.

a, RT-qPCR of GAPDH after CRISPRi treatment with two sgRNAs positioned the same distance away from the 3’ end of GAPDH as sgRNA-4 used for targeting HIDALGO (Fig. 5b). b, Comparison of HBG1 and HBG2 mature mRNA, with differences highlighted in red, a primer set that amplifies both genes equally (HBG1/2), and 3’UTR specific primer sets that uniquely amplify HBG1 or HBG2 mRNA. c, Similar to figure 6a, 3C of HIDALGO-HBG1 locus, but +/- CRISPRi (sgRNA #3) transcriptional inhibition instead of anti-sense mediated knockdown. d, Same as Figure 5e but with a primer set targeting HBG1/2 mRNA simultaneously (see above, and Supplementary Table 2) displayed with propagated error bars. e, Similar to figure 6b, calibrated ICeChIP-qPCR targeting H3K4me3 at indicated loci in either anti-sense or CRISPRi-mediated knockdown of HIDALGO compared to negative control anti-sense oligos or negative control sgRNA, or “WT” K562 cells. f, (top) Zoomed out view of the HIDALGO-HBG1-HBG2 locus, (middle) overlay of HBG1 and HBG2 gene structure with sites of primer hybridization indicated, (bottom) intron-specific primer sets in region of intron 2 for both genes HBG1 and HBG2 as well as mismatches for the cross amplification schematically depicted.

Supplementary Figure 6 Examination of early and mature HBG1/2 RNA populations during early erythroid differentiation and in response to HIDALGO depletion in K562.

a, Assessment of HBG1 and HBG2 3’UTR primer set specificity by RT-qPCR of two independent ASO (-) cDNA samples spiked with approximate indicated copies of HBG1 template corresponding to the 500bp region of HBG1 whose 3’ end is the HBG1 3’UTR (purchased from Integrated DNA Technologies). Vertical distance corresponds to log2 transformation of the fold change in 2ΔCt of spiked-in sample relative to 2ΔCt of the same cDNA sample without any spike-in; error bars correspond to standard deviation from technical qPCR reaction replicates and includes the propagated uncertainty in the zero spike-in 2ΔCt value used for fold change calculations. b, Raw mean Ct value (averaged from 3 technical replicates) vs. approximate HBG1 spike-in copy number, for samples and primers in a (linear regression fit R2 = 0.98 and 0.96). c, Same as Figure 4c, but with inclusion of RT-qPCR data from HBG1 3’UTR primer set. d, Same as c, but using the respective HBG2 instead of HBG1 primer sets. e, Same as Figure 5d, but using the respective 3’UTR primer sets instead of intron primer sets.

Supplementary Figure 7 Knockdown of known lncRNA and eRNA-associated protein factors by CRISPRi suggests alternate pathways are involved in HIDALGO regulation of HBG1.

RT-qPCR of mRNA for each target indicated, HBG1, and HIDALGO after knockdown with CRISPRi using multiple sgRNAs. Values represent 2ΔCt relative to 18S rRNA. Error bars represent S.D., n = 4 technical replicates.

Supplementary Figure 8 HIDALGO and other cheRNAs may be derived from transposable element insertion.

a, Phylogenetic comparison of HIDALGO promoter and adjacent region by MultiZ alignment (green histogram) and primate net-alignments (black bars) in the UCSC genome browser (hg38) (http://genome.ucsc.edu). b, Overlap of the indicated genomic elements with cheRNAs that have CAGE support60 (K562, n = 1895; H1, n = 358), and Gencode lncRNAs with CAGE-support compared to average random expectation derived from overlap of 1000 Monte Carlo simulations of the same spans randomly shuffled within the genome. c, phylogenetic tree of primate evolution adapted from Johnson et al. (ref. 57), highlighting the introduction of transposable elements and the relationship of fetal hemoglobin switching to simian/prosimian divergence. d, Schematic of HIDALGO/HBG1 locus and regions cloned into luciferase constructs, and normalized luciferase in K562 cells for each construct (see methods). ****P < 10-4 (Welch’s two tailed t-test).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Note 1 (PDF 2175 kb)

Supplementary Table 1 (XLSX 49 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Werner, M., Sullivan, M., Shah, R. et al. Chromatin-enriched lncRNAs can act as cell-type specific activators of proximal gene transcription. Nat Struct Mol Biol 24, 596–603 (2017). https://doi.org/10.1038/nsmb.3424

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3424

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