Comprehensive Physiology Wiley Online Library

Genome‐Wide Maps of Transcription Regulatory Elements and Transcription Enhancers in Development and Disease

Full Article on Wiley Online Library



ABSTRACT

Gene expression is regulated by numerous elements including enhancers, insulators, transcription factors, and architectural proteins. Regions of DNA distal to the transcriptional start site, called enhancers, play a central role in the temporal and tissue‐specific regulation of gene expression through RNA polymerase II. The identification of enhancers and other cis regulatory elements has largely been possible due to advances in next generation sequencing technologies. Enhancers regulate gene expression through chromatin loops mediated by architectural proteins such as YY1, CTCF, the cohesin complex, and LDB1. Additionally, enhancers can be transcribed to produce noncoding RNAs termed enhancer RNAs that likely participate in transcriptional regulation. The central role of enhancers in regulating gene expression implicates them in both normal physiology but also many disease states. The importance of enhancers is evident by the suggested role of SNPs, duplications, and other alterations of enhancer function in many diseases, ranging from cancer to atherosclerosis to chronic kidney disease. Although much progress has been made in recent years, the field of enhancer biology and our knowledge of the cis regulome remains a work in progress. This review will highlight recent seminal studies which demonstrate the role of enhancers in normal physiology and disease pathogenesis. © 2019 American Physiological Society. Compr Physiol 9:439‐455, 2019.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Trans factors involved in chromatin looping.YY1 dimerization increases chromatin looping and enhancer‐promoter interactionCohesin and CTCF are enriched at chromatin loop anchorsLDB1 is enriched at the loop anchors at the β‐globin locus
Figure 2. Figure 2. Enhancers versus promoters.Enhancers function independent of orientation. If an enhancer is flipped, it maintains its gene regulation function. A flipped promoter, however, will block gene transcription.Enhancers function independent of location. An enhancer that is moved downstream or upstream remains functional. A promoter that is no longer directly adjacent to the TSS will not drive transcription.
Figure 3. Figure 3. Characteristic histone marks.Active promoter: H3K4me3Poised enhancer: H3K4me1, H3K27me3Active enhancer: H3K4me1, H3K27acSilenced region: H3K9me2 or H3K9me3
Figure 4. Figure 4. Enhancers and RNA polymerase II. Proteins, such as Paf1, JMDJ6, BRD4, or others, bind enhancers, and promote PTEFb‐mediated phosphorylation of NELF (negative elongation factor), DSIF (DRB sensitive inducing factor) and RNA pol II. RNA pol II is released, and transcriptional elongation occurs.
Figure 5. Figure 5. eRNAsChIP‐Seq analysis identifies eRNA producing enhancers by enrichment for H3K27Ac and RNA pol IIModel of eRNA function at Nanog function. eRNAs appear to facilitate or stabilize chromatin loops between specific enhancer:gene promoter combinations.
Figure 6. Figure 6. Proposed models of alteration of enhancer function leads to cancer development.Activation of enhancers results in the opening of chromatin, leading to the greater likelihood of chromosomal rearrangements that ultimately lead to aneuploidy (21).Duplication of enhancer sequences that regulate MYC leads to increased MYC expression that drives epithelial cancers (121).Translocation of G2DHE (a GATA2 enhancer) near the EVI1 locus leads to overexpression of the EVI1 oncogene, promoting myeloid and B‐cell leukemic development (119).Heterozygous loss of cohesin leads to changes in nuclear architecture that can alter enhancer‐promoter interactions and lead to gene expression changes that promote myeloid transformation.


Figure 1. Trans factors involved in chromatin looping.YY1 dimerization increases chromatin looping and enhancer‐promoter interactionCohesin and CTCF are enriched at chromatin loop anchorsLDB1 is enriched at the loop anchors at the β‐globin locus


Figure 2. Enhancers versus promoters.Enhancers function independent of orientation. If an enhancer is flipped, it maintains its gene regulation function. A flipped promoter, however, will block gene transcription.Enhancers function independent of location. An enhancer that is moved downstream or upstream remains functional. A promoter that is no longer directly adjacent to the TSS will not drive transcription.


Figure 3. Characteristic histone marks.Active promoter: H3K4me3Poised enhancer: H3K4me1, H3K27me3Active enhancer: H3K4me1, H3K27acSilenced region: H3K9me2 or H3K9me3


Figure 4. Enhancers and RNA polymerase II. Proteins, such as Paf1, JMDJ6, BRD4, or others, bind enhancers, and promote PTEFb‐mediated phosphorylation of NELF (negative elongation factor), DSIF (DRB sensitive inducing factor) and RNA pol II. RNA pol II is released, and transcriptional elongation occurs.


Figure 5. eRNAsChIP‐Seq analysis identifies eRNA producing enhancers by enrichment for H3K27Ac and RNA pol IIModel of eRNA function at Nanog function. eRNAs appear to facilitate or stabilize chromatin loops between specific enhancer:gene promoter combinations.


Figure 6. Proposed models of alteration of enhancer function leads to cancer development.Activation of enhancers results in the opening of chromatin, leading to the greater likelihood of chromosomal rearrangements that ultimately lead to aneuploidy (21).Duplication of enhancer sequences that regulate MYC leads to increased MYC expression that drives epithelial cancers (121).Translocation of G2DHE (a GATA2 enhancer) near the EVI1 locus leads to overexpression of the EVI1 oncogene, promoting myeloid and B‐cell leukemic development (119).Heterozygous loss of cohesin leads to changes in nuclear architecture that can alter enhancer‐promoter interactions and lead to gene expression changes that promote myeloid transformation.
References
 1.Amorim S, Stathis A, Gleeson M, Iyengar S, Magarotto V, Leleu X, Morschhauser F, Karlin L, Broussais F, Rezai K, Herait P, Kahatt C, Lokiec F, Salles G, Facon T, Palumbo A, Cunningham D, Zucca E, Thieblemont C. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: A dose‐escalation, open‐label, pharmacokinetic, phase 1 study. Lancet Haematol 3(4): e196‐e204, 2016.
 2.Andersson R, Gebhard C, Miguel‐Escalada I, Hoof I, Bornholdt J, Boyd M, Chen Y, Zhao X, Schmidl C, Suzuki T, Ntini E, Arner E, Valen E, Li K, Schwarzfischer L, Glatz D, Raithel J, Lilje B, Rapin N, Bagger FO, Jørgensen M, Andersen PR, Bertin N, Rackham O, Burroughs AM, Baillie JK, Ishizu Y, Shimizu Y, Furuhata E, Maeda S, Negishi Y, Mungall CJ, Meehan TF, Lassman T, Itoh M, Kawahi H, Kondo N, Kawai J, Lennartsson A, Daub CO, Heutink P, Hume DA, Jensen TH, Suzuki H, Hayashizaki Y, Müller F, Forrest ARR, Carninci P, Rehli M, Sandelin A. An atlas of active enhancers across human cell types and tissues. Nature 507(7493): 455‐461, 2014.
 3.Arnold CD, Gerlach D, Stelzer C, Boryn LM, Rath M, Stark A. Genome‐wide quantitative enhancer activity maps identified by STARR‐seq. Science 339(6123): 1074‐1077, 2013.
 4.Arnosti DN, Barolo S, Levine M, Small S. The eve stripe 2 enhancer employs multiple modes of transcriptional synergy. Development 122(1): 205‐214, 1996.
 5.Arzate‐Mejía RG, Recillas‐Targa F, Corces VG. Developing in 3D: The role of CTCF in cell differentiation. Development 145(6): pii: dev137729, 2018.
 6.Banerji J, Olson L, Schaffner W. A lymphocyte‐specific cellular enhancer is located downstream of the joining region in immunoglobulin heavy chain genes. Cell 33(3): 729‐740, 1983.
 7.Banerji J, Rusconi S, Schaffner W. 1981. Expression of a β‐globin gene is enhanced by remote SV40 DNA sequences. Cell 27(2): 299‐308, 1981.
 8.Barski A, Cuddapah S, Cui K, Roh T‐Y, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K. High‐resolution profiling of histone methylations in the human gsenome. Cell 129(4): 823‐837, 2007.
 9.Berlivet S, Paquette D, Dumouchel A, Langlais D, Dostie J, Kmita M. Clustering of tissue‐specific sub‐TADs accompanies the regulation of HoxA genes in developing limbs. PLoS Genet 9(12): e1004018, 2013.
 10.Bernstein BE, Kamal M, Lindblad‐Toh K, Bekiranov S, Bailey DK, Huebert DJ, McMahon S, Karlsson EK, Kulbokas EJ, III, Gingeras TR, Schreiber SL, Lander ES. Genomic maps and comparative analysis of histone modifications in human and mouse. Cell 120(2): 169‐181, 2005.
 11.Berthon C, Raffoux E, Thomas X, Vey N, Gomez‐Roca C, Yee K, Taussig DC, Rezai K, Roumier C, Herait P, Kahatt C, Quesnel B, Michallet M, Recher C, Lokiec F, Preudhomme C. Bromodomain inhibitor OTX015 in patients with acute leukaemia: A dose‐escalation, phase 1 study. Lancet Haematol 3(4): e186‐e195, 2016.
 12.Blinka S, Reimer MH, Jr., Pulakanti K, Rao S. Super‐Enhancers at the Nanog locus differentially regulate neighboring pluripotency‐associated genes. Cell Rep 17(1): 19‐28, 2016.
 13.Brand M, Ranish JA, Kummer NT, Hamilton J, Igarashi K, Francastel C, Chi TH, Crabtree GR, Aebersold R, Groudine M. Dynamic changes in transcription factor complexes during erythroid differentiation revealed by quantitative proteomics. Nat Struct Mol Biol 11(1): 73‐80, 2004.
 14.Brandt MM, Meddens CA, Louzao‐Martinez L, van den Dungen NAM, Lansu NR, Nieuwenhuis EES, Duncker DJ, Verhaar MC, Joles JA. Mokry M, Cheng C. Chromatin conformation links distal target genes to CKD loci. J Am Soc Nephrol 29(2): 462‐476, 2018.
 15.Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA‐binding proteins and nucleosome position. Nat Methods 10(12): 1213‐18, 2013.
 16.Bulger M, Groudine M. Looping versus linking: Toward a model for long‐distance gene activation. Genes Dev 13(19): 2465‐2477, 1999.
 17.Cancer Genome Atlas Research Network, Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, Robertson A, Hoadley K, Triche TJ, Jr., Laird PW, Baty JD, Fulton LL, Fulton R, Heath SE, Kalicki‐Veizer J, Kandoth C, Klco JM, Koboldt DC, Kanchi KL, Kulkarni S, Lamprecht TL, Larson DE, Lin L, Lu C, McLellan MD, McMichael JF, Payton J, Schmidt H, Spencer DH, Tomasson MH, Wallis JW, Wartman LD, Watson MA, Welch J, Wendl MC, Ally A, Balasundaram M, Birol I, Butterfield Y, Chiu R, Chu A, Chuah E, Chun HJ, Corbett R, Dhalla N, Guin R, He A, Hirst C, Hirst M, Holt RA, Jones S, Karsan A, Lee D, Li HI, Marra MA, Mayo M, Moore RA, Mungall K, Parker J, Pleasance E, Plettner P, Schein J, Stoll D, Swanson L, Tam A, Thiessen N, Varhol R, Wye N, Zhao Y, Gabriel S, Getz G, Sougnez C, Zou L, Leiserson MD, Vandin F, Wu HT, Applebaum F, Baylin SB, Akbani R, Broom BM, Chen K, Motter TC, Nguyen K, Weinstein JN, Zhang N, Ferguson ML, Adams C, Black A, Bowen J, Gastier‐Foster J, Grossman T, Lichtenberg T, Wise L, Davidsen T, Demchok JA, Shaw KR, Sheth M, Sofia HJ, Yang L, Downing JR, Eley G. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 368(22): 2059‐2074, 2013.
 18.Canver MC, Smith EC, Sher F, Pinello L, Sanjana NE, Shalem O, Chen DD, Schupp PG, Vinjamur DS, Garcia SP, Luc S, Kurita R, Nakamura Y, Fujiwara Y, Maeda T, Yuan GC, Zhang F, Orkin SH, Bauer DE. BCL11A enhancer dissection by Cas9‐mediated in situ saturating mutagenesis. Nature 527(7577): 192‐197, 2015.
 19.Chatterjee S, Ahituv N. Gene regulatory elements, major drivers of human disease. Annu Rev Genomics Hum Genet 18: 45‐63, 2017.
 20.Chen FX, Woodfin AR, Gardini A, Rickels RA, Marshall SA, Smith ER, Shiekhattar R, Shilatifard A. PAF1, a molecular regulator of promoter‐proximal pausing by RNA polymerase II. Cell 162(5): 1003‐1015, 2015.
 21.Chen H, Li C, Peng X, Zhou Z, Weinstein JN, Cancer Genome Atlas Research Network, Liang H. A Pan‐cancer analysis of enhancer expression in nearly 9000 patient samples. Cell 173(2): 386‐399.e12, 2018.
 22.Core LJ, Waterfall JJ, Lis JT. Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science 322(5909): 1845‐1848, 2008.
 23.Cotney J, Leng J, Oh S, DeMare LE, Reilly SK, Gerstein MB, Noonan JP. Chromatin state signatures associated with tissue‐specific gene expression and enhancer activity in the embryonic limb. Genome Res 22(6): 1069‐1080, 2012.
 24.Das S, Senapati P, Chen Z, Reddy MA, Ganguly R, Lanting L, Mandi V, Bansal A, Leung A, Zhang S, Jia Y, Wu X, Schones DE, Natarajan R. Regulation of angiotensin II actions by enhancers and super‐enhancers in vascular smooth muscle cells. Nat Commun 8(1): 1467, 2017.
 25.Dekker J. The three “C” s of chromosome conformation capture: Controls, controls, controls. Nat Methods 3(1): 17‐21, 2006.
 26.de Laat W, Duboule D. Topology of mammalian developmental enhancers and their regulatory landscapes. Nature 502(7472): 499‐506, 2013.
 27.Deng W, Lee J, Wang H, Miller J, Reik A, Gregory PD, Dean A, Blobel GA. Controlling long‐range genomic interactions at a native locus by targeted tethering of a looping factor. Cell 149(6): 1233‐1244, 2012.
 28.Deng W, Rupon JW, Krivega I, Breda L, Motta I, Jahn KS, Reik A, Gregory PD, Rivella S, Dean A, Blobel GA. Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell 158(4): 849‐860, 2014.
 29.De Santa F, Barozzi I, Mietton F, Ghisletti S, Polletti S, Tusi BK, Muller H, Ragoussis J, Wei CL, Natoli G. A large fraction of extragenic RNA pol II transcription sites overlap enhancers. PLoS Biol 8(5): e1000384, 2010.
 30.Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, Hu M, Liu JS, Ren B. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485(7398): 376‐380, 2012.
 31.Donohoe ME, Zhang X, McGinnis L, Biggers J, Li E, Shi Y. Targeted disruption of mouse Yin Yang 1 transcription factor results in peri‐implantation lethality. Mol Cell Biol 19(10): 7237‐7244, 1999.
 32.Dowen JM, Fan ZP, Hnisz D, Ren G, Abraham BJ, Zhang LN, Weintraub AS, Schujiers J, Lee TI, Zhao K, Young RA. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell 159(2): 374‐387, 2014.
 33.Dukler N, Gulko B, Huang Y‐F, Siepel A. Is a super‐enhancer greater than the sum of its parts? Nat Genet 49(1): 2‐3, 2016.
 34.Elgin SC. The formation and function of DNase I hypersensitive sites in the process of gene activation. J Biol Chem 263(36): 19259‐1962, 1988.
 35.Emilsson V, Thorleifsson G, Zhang B, Leonardson AS, Zink F, Zhu J, Carlson S, Helgason A, Walters GB, Gunnarsdottir S, Mouy M, Steinthorsdottir V, Eiriksdottir GH, Bjornsdottir G, Reynisdottir I, Gudbjartsson D, Helgadottir A, Jonasdottir A, Jonasdottir A, Styrkarsdottir U, Gretarsdottir S, Magnusson KP, Stefansson H, Fossdal R, Kristjansson K, Gislason HG, Stefansson T, Leifsson BG, Thorsteinsdottir U, Lamb JR, Gulcher JR, Reitman ML, Kong A, Schadt EE, Stefansson K. Genetics of gene expression and its effect on disease. Nature 452(7186): 423‐428, 2008.
 36.Ferrier P, Krippl B, Blackwell TK, Furley AJ, Suh H, Winoto A, Cook WD, Hood L, Constantini F, Alt FW. Separate elements control DJ and VDJ rearrangement in a transgenic recombination substrate. EMBO J 9(1): 117‐125, 1990.
 37.Fisher JB, Peterson J, Reimer M, Stelloh C, Pulakanti K, Gerbec ZJ, Strouse JM, Strouse C, McNulty M, Malarkannan S, Crispino JD, Milanovich S, Rao, S. The cohesin subunit Rad21 is a negative regulator of hematopoietic self‐renewal through epigenetic repression of Hoxa7 and Hoxa9. Leukemia 31(3): 712‐719, 2017.
 38.Fong CY, Gilan O, Lam EYN, Rubin AF, Ftouni S, Tyler D, Stanlley K, Sinha D, Yeh P, Morison J, Giotopoulos G, Lugo D, Jeffrey P, Lee SC, Carpenter C, Gregory R, Ramsay RG, Lane SW, Abdel‐Wahab O, Kouzarides T, Johnstone RW, Dawson SJ, Huntly BJ, Prinjha RK, Papenfuss AT, Dawson MA. BET inhibitor resistance emerges from leukaemia stem cells. Nature 525(7570): 538‐542, 2015.
 39.Fulco CP, Munschauer M, Anyoha R, Munson G, Grossman SR, Perez EM, Kane M, Cleary B, Lander ES, Engreitz JM. Systematic mapping of functional enhancer‐promoter connections with CRISPR interference. Science 354(6313): 769‐773, 2016.
 40.Gavrilov A, Eivazova E, Priozhkova I, Lipinski M, Razin S, Vassetzky Y. Chromosome conformation capture (from 3C to 5C) and its ChIP‐based modification. Methods Mol Biol 567: 171‐188, 2009.
 41.Ghavi‐Helm Y, Klein FA, Pakozdi T, Ciglar L, Noordermeer D, Huber W, Furlong EE. Enhancer loops appear stable during development and are associated with paused polymerase. Nature 512(7512): 96‐100, 2014.
 42.Gillies SD, Morrison SL, Oi VT, Tonegawa S. A tissue‐specific transcription enhancer element is located in the major intron of a rearranged immunoglobulin heavy chain gene. Cell 33(3): 717‐728, 1983.
 43.Heintzman ND, Hon GC, Hawkins RD, Kheradpour P, Stark A, Harp LF, Ye Z, Lee LK, Stuart RK, Ching CW, Ching KA, Antosiewicz‐Bourget JE, Liu H, Zhang X, Green RD, Lobanenkov VV, Stewart R, Thomson JA, Crawford GE, Kellis M, Ren B. Histone modifications at human enhancers reflect global cell‐type‐specific gene expression. Nature 459(7243): 108‐112, 2009.
 44.Heintzman ND, Stuart RK, Hon G, Fu Y, Ching CW, Hawkins RD, Barrera LO, Van Calcar S, Qu C, Ching KA, Wang W, Weng Z, Green RD, Crawford GE, Ren B. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet 39(3): 311‐318, 2007.
 45.Hnisz D, Abraham BJ, Lee TI, Lau A, Saint‐André V, Sigova AA, Hoke HA, Young RA. Super‐enhancers in the control of cell identity and disease. Cell 155(4): 934‐947, 2013.
 46.Hnisz D, Day DS, Young RA. Insulated neighborhoods: Structural and functional units of mammalian gene control. Cell 167(5): 1188‐1200, 2016.
 47.Hori R, Carey M. The role of activators in assembly of RNA polymerase II transcription complexes. Curr Opin Genet Dev 4(2): 236‐244, 1994.
 48.Ing‐Simmons E, Seitan VC, Faure AJ, Flicek P, Carroll T, Dekker J, Risher AG, Lenhard B, Merkenschlager M. Spatial enhancer clustering and regulation of enhancer‐proximal genes by cohesin. Genome Res 25(4): 504‐513, 2015.
 49.Jin F, Li Y, Dixon JR, Selvaraj S, Ye Z, Lee AY, Yen C‐A, Schmitt AD, Espinoza CA, Ren B. A high‐resolution map of the three‐dimensional chromatin interactome in human cells. Nature 503(7475): 290‐294, 2013.
 50.Kagey MH, Newman JJ, Bilodeau S, Zhan Y, Orlando DA, van Berkum NL, Ebmeier CC, Goossens J, Rahl PB, Levine SS, Taatjes DJ, Dekker J, Young RA. Mediator and cohesin connect gene expression and chromatin architecture. Nature 467(7314): 430‐435, 2010.
 51.Kim T‐K, Hemberg M, Gray JM, Costa AM, Bear DM, Wu J, Harmin DA, Laptewicz M, Barbara‐Haley K, Kuersten S, Markenscoff‐Papadimitriou E, Kuhl D, Bito H, Worley PF, Kreiman G, Greenberg ME. Widespread transcription at neuronal activity‐regulated enhancers. Nature 465(7295): 182‐187, 2010.
 52.Kioussis D, Vanin E, deLange T, Flavell RA, Grosveld FG. Beta‐globin gene inactivation by DNA translocation in gamma beta‐thalassaemia. Nature 306(5944): 662‐666, 1983.
 53.Krivega I, Dean A. LDB1‐mediated enhancer looping can be established independent of mediator and cohesin. Nucleic Acids Res 45(14): 8255‐8268, 2017.
 54.Kumar K, Raza SS, Knab LM, Chow CR, Kwok B, Bentrem DJ, Popovic R, Ebine K, Licht JD, Munshi HG. GLI2‐dependent c‐MYC upregulation mediates resistance of pancreatic cancer cells to the BET bromodomain inhibitor JQ1. Sci Rep 5: 9489, 2015.
 55.Leeke B, Marsman J, O'Sullivan JM, Horsfield JA. Cohesin mutations in myeloid malignancies: Underlying mechanisms. Exp Hematol Oncol 3: 13, 2014.
 56.Li L, Jothi R, Cui K, Lee JY, Cohen T, Gorivodsky M, Tzchori I, Zhao Y, Hayes SM, Bresnick EH, Zhao K, Westphal H, Love PE. Nuclear adaptor Ldb1 regulates a transcriptional program essential for the maintenance of hematopoietic stem cells. Nat Immunol 12(2): 129‐136, 2011.
 57.Liu W, Ma Q, Wong K, Li W, Ohgi K, Zhang J, Aggarwal A, Rosenfeld MG. Brd4 and JMJD6‐associated anti‐pause enhancers in regulation of transcriptional pause release. Cell 155(7): 1581‐1595, 2013.
 58.Liu X, Zhang Y, Chen Y, Li M, Zhou F, Li K, Cao H, Ni M, Liu Y, Gu Z, Dickerson KE, Xie S, Hon GC, Xuan Z, Zhang MQ, Shao Z, Xu J. In situ capture of chromatin interactions by biotinylated dCas9. Cell 170(5): 1028‐1043.e19, 2017.
 59.Li W, Notani D, Ma Q, Tanasa B, Nunez E, Chen AY, Merkurjev D, Zhang J, Ohgi K, Song X, Oh S, Kim HS, Glass CK, Rosenfeld MG. Functional roles of enhancer RNAs for oestrogen‐dependent transcriptional activation. Nature 498(7455): 516‐520, 2013.
 60.Li W, Notani D, Rosenfeld MG. Enhancers as non‐coding RNA transcription units: Recent insights and future perspectives. Nat Rev Genet 17(4): 207‐223, 2016.
 61.Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, Bradner JE, Lee TI, Young RA. Selective inhibition of tumor oncogenes by disruption of super‐enhancers. Cell 153(2): 320‐334, 2013.
 62.Mitter D, Chiaie BD, Lüdecke H‐J, Gillessen‐Kaesbach G, Bohring A, Kohlhase J, Caliebe A, Siebert R, Roepke A, Ramos‐Arroyo MA, Nieva B, Menten B, Loeys B, Mortier G, Wieczorek D. Genotype‐phenotype correlation in eight new patients with a deletion encompassing 2q31.1. Am J Med Genet A 152A(5): 1213‐1224, 2010.
 63.Montavon T, Soshnikova N, Mascrez B, Joye E, Thevenet L, Splinter E, de Laat W, Spitz F, Duboule D. A regulatory archipelago controls hox genes transcription in digits. Cell 147(5): 1132‐1145, 2011.
 64.Moorthy SD, Davidson S, Shchuka VM, Singh G, Malek‐Gilani N, Langroudi L, Martchenko A, So V, Macpherson NN, Mitchell JA. Enhancers and super‐enhancers have an equivalent regulatory role in embryonic stem cells through regulation of single or multiple genes. Genome Res 27(2): 246‐258, 2017.
 65.Moreau P, Hen R, Wasylyk B, Everett R, Gaub MP, Chambon P. The SV40 72 base repair repeat has a striking effect on gene expression both in SV40 and other chimeric recombinants. Nucleic Acids Res 9(22): 6047‐6068, 1981.
 66.Mousavi K, Zare H, Dell'orso S, Grontved L, Gutierrez‐Cruz G, Derfoul A, Hager GL, Sartorelli V. eRNAs promote transcription by establishing chromatin accessibility at defined genomic loci. Mol Cell 51(5): 606‐617, 2013.
 67.Mullenders J, Aranda‐Orgilles B, Lhoumaud P, Keller M, Pae J, Wang K, Kayembe C, Rocha PP, Raviram R, Gong Y, Premsrirut PK, Tsirigos A, Bonneau R, Skok JA, Cimmino L, Hoehn D, Aifantis I. Cohesin loss alters adult hematopoietic stem cell homeostasis, leading to myeloproliferative neoplasms. J Exp Med 212(11): 1833‐1850, 2015.
 68.Murakawa Y, Yoshihara M, Kawaji H, Nishikawa M, Zayed H, Suzuki H, Fantom C, Hayashizaki Y. Enhanced identification of transcriptional enhancers provides mechanistic insights into diseases. Trends Genet 32(2): 76‐88, 2016.
 69.Muse GW, Gilchrist DA, Nechaev S, Shah R, Parker JS, Grissom SF, Zeitlinger J, Adelman K. RNA polymerase is poised for activation across the genome. Nat Genet 39(12): 1507‐1511, 2007.
 70.Narendra V, Rocha PP, An D, Raviram R, Skok JA, Mazzoni EO, Reinberg D. CTCF establishes discrete functional chromatin domains at the Hox clusters during differentiation. Science 347(6225): 1017‐1021, 2015.
 71.Nicholls SJ, Puri R, Wolski K, Ballantyne CM, Barter PJ, Brewer HB, Kastelein JJ, Hu B, Uno K, Kataoka Y, Herrman JP, Merkely B, Borgman M, Nissen SE. Effect of the BET protein inhibitor, RVX‐208, on progression of coronary atherosclerosis: Results of the phase 2b, randomized, double‐blind, multicenter, ASSURE trial. Am J Cardiovasc Drugs 16(1): 55‐65, 2015.
 72.Noordermeer D, Leleu M, Schorderet P, Joye E, Chabaud F, Duboule D. Temporal dynamics and developmental memory of 3D chromatin architecture at Hox gene loci. Elife 3: e02557, 2014.
 73.Noordermeer D, Leleu M, Splinter E, Rougemont J, De Laat W, Duboule D. The dynamic architecture of Hox gene clusters. Science 334(6053): 222‐225, 2011.
 74.Nora EP, Goloborodko A, Valton A‐L, Gibcus JH, Uebersohn A, Abdennur N, Dekker J, Mirny LA, Bruneau BG. Targeted degradation of CTCF decouples local insulation of chromosome domains from genomic compartmentalization. Cell 169(5): 930‐944.e22, 2017.
 75.Nora EP, Lajoie BR, Schulz EG, Giorgetti L, Okamoto I, Servant N, Piolot T, van Berkum NL, Meisig J, Sedat J, Gribnau J, Barillot E, Blüthgen N, Dekker J, Heard E. Spatial partitioning of the regulatory landscape of the X‐inactivation centre. Nature 485(7398): 381‐385, 2012.
 76.Ohkura H, Adachi Y, Kinoshita N, Niwa O, Toda T, Yanagida M. Cold‐sensitive and caffeine‐supersensitive mutants of the Schizosaccharomyces pombe dis genes implicated in sister chromatid separation during mitosis. EMBO J 7(5): 1465‐1473, 1988.
 77.O'Kane CJ, Gehring WJ. Detection in situ of genomic regulatory elements in Drosophila. Proc Natl Acad Sci 84(24): 9123‐9127, 1987.
 78.Ong C‐T, Corces VG. CTCF: An architectural protein bridging genome topology and function. Nat Rev Genet 15(4): 234‐246, 2014.
 79.Ørom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, Lai F, Zytnicki M, Notredame C, Huang Q, Guigo R, Shiekhattar R. Long noncoding RNAs with enhancer‐like function in human cells. Cell 143(1): 46‐58, 2010.
 80.Parelho V, Hadjur S, Spivakov M, Leleu M, Sauer S, Gregson HC, Jarmuz A, Canzonetta C, Webster Z, Nesterova T, Cobb BS, Yokomori K, Dillon N, Aragon L, Fisher AG, Merkenschlager M. Cohesins functionally associate with CTCF on mammalian chromosome arms. Cell 132(3): 422‐433, 2008.
 81.Peeters JGC, Vervoort SJ, Tan SC, Mijnheer G, de Roock S, Vastert SJ, Nieuwenhuis EE, van Wijk F, Prakken BJ, Creyghton MP, Coffer PJ, Mokry M, van Loosdregt J. Inhibition of super‐enhancer activity in autoinflammatory site‐derived T cells reduces disease‐associated gene expression. Cell Rep 12(12): 1986‐1996, 2015.
 82.Pott S, Lieb JD. What are super‐enhancers? Nat Genet 47(1): 8‐12, 2015.
 83.Pulakanti K, Pinello L, Stelloh C, Blinka S, Allred J, Milanovich S, Kiblawi S, Peterson J, Wang A, Yuan GC, Rao S. Enhancer transcribed RNAs arise from hypomethylated, Tet‐occupied genomic regions. Epigenetics 8(12): 1303‐1320, 2013.
 84.Rada‐Iglesias A, Bajpai R, Swigut T, Brugmann SA, Flynn RA, Wysocka J. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470(7333): 279‐283, 2011.
 85.Rao S, Huang S‐C, St. Hilaire BG, Engreitz JM, Perez EM, Kieffer‐Kwon KR, Sanborn AL, Johnstone SE, Bascom GD, Bochkov ID, Huang X, Shamim MS, Shin J, Turner D, Ye Z, Omer AD, Robinson JT, Schlick T, Bernstein BE, Casellas R, Lander ES, Aiden EL. Cohesin loss eliminates all loop domains, leading to links among superenhancers and downregulation of nearby genes. Cell 171(2): 305‐320 2017.
 86.Remeseiro S, Losada A. Cohesin, a chromatin engagement ring. Curr Opin Cell Biol 25(1): 63‐71, 2013.
 87.Ren G, Jin W, Cui K, Rodrigez J, Hu G, Zhang Z, Larson DR, Zhao K. CTCF‐mediated enhancer‐promoter interaction is a critical regulator of cell‐to‐cell variation of gene expression. Mol Cell 67(6): 1049‐1058.e6, 2017.
 88.Rippe K, von Hippel PH, Langowski J. Action at a distance: DNA‐looping and initiation of transcription. Trends Biochem Sci 20(12): 500‐506, 1995.
 89.Sati S, Cavalli G. Chromosome conformation capture technologies and their impact in understanding genome function. Chromosoma 126(1): 33‐44, 2017.
 90.Sawado T, Halow J, Bender MA, Groudine M. The beta‐globin locus control region (LCR) functions primarily by enhancing the transition from transcription initiation to elongation. Genes Dev 17(8): 1009‐1018, 2003.
 91.Schaukowitch K, Joo J‐Y, Liu X, Watts JK, Martinez C, Kim T‐K. Enhancer RNA facilitates NELF release from immediate early genes. Mol Cell 56(1): 29‐42, 2014.
 92.Schwarzer W, Abdennur N, Goloborodko A, Pekowska A, Fudenberg G, Loe‐Mie Y, Fonseca NA, Huber W, H Haering C, Mirny L, Spitz F. Two independent modes of chromatin organization revealed by cohesin removal. Nature 551(7678): 51‐56, 2017.
 93.Seitan VC, Faure AJ, Zhan Y, McCord RP, Lajoie BR, Ing‐Simmons E, Lenhard B, Giorgetti L, Heard E, Fisher AG, Flicek P, Dekker J, Merkenschlager M. Cohesin‐based chromatin interactions enable regulated gene expression within preexisting architectural compartments. Genome Res 23(12): 2066‐2077, 2013.
 94.Siebel AL, Trinh SK, Formosa MF, Mundra PA, Natoli AK, Reddy‐Luthmoodoo M, Huynh K, Khan AA, Carey AL, van Hall G, Cobelli C, Dalla‐Man C, Otvos JD, Rye KA, Johansson J, Gordon A, Wong NC, Sviridov D, Barter P, Duffy SJ, Meikle PJ, Kingwell BA. Effects of the BET‐inhibitor, RVX‐208 on the HDL lipidome and glucose metabolism in individuals with prediabetes: A randomized controlled trial. Metabolism 65(6): 904‐914, 2016.
 95.Sigova AA, Abraham BJ, Ji X, Molinie B, Hannett NM, Guo YE, Jangi M, Giallourakis CC, Sharp PA, Young RA. Transcription factor trapping by RNA in gene regulatory elements. Science 350(6263): 978‐981, 2015.
 96.Sofueva S, Yaffe E, Chan W‐C, Georgopoulou D, Vietri Rudan M, Mira‐Bontenbal H, Pollard SM, Schroth GP, Tanay A, Hadjur S. Cohesin‐mediated interactions organize chromosomal domain architecture. EMBO J 32(24): 3119–3129, 2013.
 97.Song S‐H, Hou C, Dean A. A positive role for NLI/Ldb1 in long‐range β‐globin locus control region function. Mol Cell 28(5): 810‐822, 2007.
 98.Stadler MB, Murr R, Burger L, Ivanek R, Lienert F, Schöler A, van Nimwegen E, Wirbelauer C, Oakeley EJ, Gaidatzis D, Tiwari VK, Schübeler D. DNA‐binding factors shape the mouse methylome at distal regulatory regions. Nature 480(7378): 490‐495, 2011.
 99.Stanojevic D, Small S, Levine M. Regulation of a segmentation stripe by overlapping activators and repressors in the Drosophila embryo. Science 254(5036): 1385‐1387, 1991.
 100.Stranger BE, Nica AC, Forrest MS, Dimas A, Bird CP, Beazley C, Ingle CE, Dunning M, Flicek P, Koller D, Montgomery S, Tavaré S, Deloukas P, Dermitzakis ET. Population genomics of human gene expression. Nat Genet 39(10): 1217‐1224, 2007.
 101.Tolhuis B, Palstra RJ, Splinter E, Grosveld F, de Laat W. Looping and interaction between hypersensitive sites in the active beta‐globin locus. Mol Cell 10(6): 1453‐1465, 2002.
 102.Tuan D, Kong S, Hu K. Transcription of the hypersensitive site HS2 enhancer in erythroid cells. Proc Natl Acad Sci 89(23): 11219‐11223, 1992.
 103.Vanhille L, Griffon A, Maqbool MA, Zacarias‐Cabeza J, Dao LTM, Fernandez N, Ballester B, Andrau JC, Spicuglia S. High‐throughput and quantitative assessment of enhancer activity in mammals by CapStarr‐seq. Nat Commun 6: 6905, 2015.
 104.van Steensel B, Dekker J. Genomics tools for unraveling chromosome architecture. Nat Biotechnol 28(10): 1089‐1095, 2010.
 105.Viny AD, Ott CJ, Spitzer B, Rivas M, Meydan C, Papalexi E, Yelin D, Shank K, Reyes J, Chiu A, Romin Y, Boyko V, Thota S, Maciejewski JP, Melnick A, Bradner JE, Levine RL. Dose‐dependent role of the cohesin complex in normal and malignant hematopoiesis. J Exp Med 212(11): 1819‐1832, 2015.
 106.Visel A, Blow MJ, Li Z, Zhang T, Akiyama JA, Holt A, Plajzer‐Frick I, Shoukry M, Wright C, Chen F, Afzal V, Ren B, Rubin EM, Pennacchio LA. ChIP‐seq accurately predicts tissue‐specific activity of enhancers. Nature 457(7231): 854‐858, 2009.
 107.Wadman IA, Osada H, Grütz GG, Agulnick AD, Westphal H, Forster A, Rabbitts TH. The LIM‐only protein Lmo2 is a bridging molecule assembling an erythroid, DNA‐binding complex which includes the TAL1, E47, GATA‐1 and Ldb1/NLI proteins. EMBO J 16(11): 3145‐3157, 1997.
 108.Wang J, Giaever G. Action at a distance along a DNA. Science 240(4850): 300‐304, 1988.
 109.Wang J, Wu X, Wei C, Huang X, Ma Q, Huang X, Faiola F, Guallar D, Fidalgo M, Huang T, Peng D, Chen L, Yu H, Li X, Sun J, Liu X, Cai X, Chen X, Wang L, Ren J, Wang J, Ding J. YY1 positively regulates transcription by targeting promoters and super‐enhancers through the BAF complex in embryonic stem cells. Stem Cell Reports 10(4): 1324‐1339, 2018.
 110.Wang Q, Carroll JS, Brown M. Spatial and temporal recruitment of androgen receptor and its coactivators involves chromosomal looping and polymerase tracking. Mol Cell 19(5): 631‐642, 2005.
 111.Weintraub AS, Li CH, Zamudio AV, Sigova AA, Hannett NM, Day DS, Abraham BJ, Cohen MA, Nabet B, Buckley DL, Guo YE, Hnisz D, Jaenisch R, Bradner JE, Gray NS, Young RA. YY1 is a structural regulator of enhancer‐promoter loops. Cell 171(7): 1573‐1588.e28, 2017.
 112.Whyte WA, Orlando DA, Hnisz D, Abraham BJ, Lin CY, Kagey MH, Rahl PB, Lee TI, Young RA. Master transcription factors and mediator establish super‐enhancers at key cell identity genes. Cell 153(2): 307‐319, 2013.
 113.Wiench M, John S, Baek S, Johnson TA, Sung M‐H, Escobar T, Simmons CA, Pearce KH, Biddie SC, Sabo PJ, Thurman RE, Stamatoyannopoulos JA, Hager GL. DNA methylation status predicts cell type‐specific enhancer activity. EMBO J 30(15): 3028‐3039, 2011.
 114.Williamson I, Hill RE, Bickmore WA. Enhancers: From developmental genetics to the genetics of common human disease. Dev Cell 21(1): 17‐19, 2011.
 115.Willi M, Yoo KH, Reinisch F, Kuhns TM, Lee HK, Wang C, Hennighausen L. Facultative CTCF sites moderate mammary super‐enhancer activity and regulate juxtaposed gene in non‐mammary cells. Nat Commun 8: 16069, 2017.
 116.Willi M, Yoo KH, Wang C, Trajanoski Z, Hennighausen L. Differential cytokine sensitivities of STAT5‐dependent enhancers rely on Stat5 autoregulation. Nucleic Acids Res 44(21): 10277‐10291, 2016.
 117.Wu H, Nord AS, Akiyama JA, Shoukry M, Afzal V, Rubin EM, Pennacchio LA, Visel A. Tissue‐specific RNA expression marks distant‐acting developmental enhancers. PLoS Genet 10(9): e1004610, 2014.
 118.Wutz G, Várnai C, Nagasaka K, Cisneros DA, Stocsits RR, Tang W, Schoenfelder S, Jessberger G, Muhar M, Hossain MJ, Walther N, Koch B, Kueblbeck M, Ellenberg J, Zuber J, Fraser P, Peters JM. Topologically associating domains and chromatin loops depend on cohesin and are regulated by CTCF, WAPL, and PDS5 proteins. EMBO J 36(24): 3573‐3599, 2017.
 119.Yamazaki H, Suzuki M, Otsuki A, Shimizu R, Bresnick EH, Engel JD, Yamamoto M. A remote GATA2 hematopoietic enhancer drives leukemogenesis in inv(3)(q21;q26) by activating EVI1 expression. Cancer Cell 25(4): 415‐427, 2014.
 120.Yu M, Yang W, Ni T, Tang Z, Nakadai T, Zhu J, Roeder RG. RNA polymerase II‐associated factor 1 regulates the release and phosphorylation of paused RNA polymerase II. Science 350(6266): 1383‐1386, 2015.
 121.Zhang X, Choi PS, Francis JM, Imielinski M, Watanabe H, Cherniack AD, Meyerson M. Identification of focally amplified lineage‐specific super‐enhancers in human epithelial cancers. Nat Genet 48(2): 176‐182, 2016.
 122.Zhao Z, Tavoosidana G, Sjölinder M, Göndör A, Mariano P, Wang S, Kanduri C, Lezcano M, Sandhu KS, Singh U, Pant V, Tiwari V, Kurukuti S, Ohlsson R. Circular chromosome conformation capture (4C) uncovers extensive networks of epigenetically regulated intra‐ and interchromosomal interactions. Nat Genet 38(11): 1341‐1347, 2006.
 123.Zhou HY, Katsman Y, Dhaliwal NK, Davidson S, Macpherson NN, Sakthidevi M, Collura F, Mitchell JA. A Sox2 distal enhancer cluster regulates embryonic stem cell differentiation potential. Genes Dev 28(24): 2699‐2711, 2014.
 124.Zuin J, Dixon JR, van der Reijden MI, Ye Z, Kolovos P, Brouwer RW, van de Corput MP, van de Werken HJ, Knoch TA, van IJcken WF, Grosveld FG, Ren B, Wendt KS. Cohesin and CTCF differentially affect chromatin architecture and gene expression in human cells. Proc Natl Acad Sci U S A 111(3): 996‐1001, 2014.

 

Teaching Material

P. Agrawal, K. E. Heimbruch, S. Rao. Genome-Wide Maps of Transcription Regulatory Elements and Transcription Enhancers in Development and Disease. Compr Physiol 9: 2019, 469-485.

Didactic Synopsis

Major Teaching Points:

  • Transcriptional regulation is a complex process controlled by numerous elements, which act either in cis or trans.
  • Enhancers are DNA elements containing transcription factor binding motifs that play an important role in transcriptional regulation in a distance and orientation independent manner.
  • Technological advances have been essential in identifying enhancers and their role in regulating gene expression.
  • Enhancers regulate gene expression through interactions with RNA polymerase II by regulating transcriptional elongation.
  • Enhancers operate by being in close physical proximity to the genes they regulate within the nucleus. This is mediated by chromatin looping mediated by architectural proteins such as YY1, CTCF, LDB1, and the cohesin complex.
  • SNPs, duplications, and other alterations in enhancer function have been implicated in a wide variety of diseases.

Didactic Legends

The figures—in a freely downloadable PowerPoint format—can be found on the Images tab along with the formal legends published in the article. The following legends to the same figures are written to be useful for teaching.

Figure 1 Trans factors involved in chromatin looping. (A) YY1 dimerization increases chromatin looping and enhancer-promoter interaction (B) Cohesin and CTCF are enriched at chromatin loop anchors (C) LDB1 is enriched at the loop anchors at the β-globin locus Teaching point: Chromatin looping may be due to multiple different mechanisms

Figure 2 Enhancers versus promoters. (A) Enhancers function independent of orientation. If an enhancer is flipped, it maintains its gene regulation function. A flipped promoter, however, will block gene transcription. (B) Enhancers function independent of location. An enhancer that is moved downstream or upstream remains functional. A promoter that is no longer directly adjacent to the TSS will not drive transcription. Teaching point: Enhancers function independent of orientation and location, unlike promoters.

Figure 3 Characteristic histone marks. (A) Active promoter: H3K4me3 (B) Poised enhancer: H3K4me1, H3K27me3 (C) Active enhancer: H3K4me1, H3K27ac (D) Silenced region: H3K9me2 or H3K9me3 Teaching point: the activity or lack thereof of enhancers can be inferred by various histone marks.

Figure 4 Enhancers and RNA polymerase II. Proteins, such as Paf1, JMDJ6, BRD4 or others, bind enhancers, and promote PTEFb-mediated phosphorylation of NELF (negative elongation factor), DSIF (DRB sensitive inducing factor) and RNA pol II. RNA pol II is released, and transcriptional elongation occurs. Teaching point: Enhancers interact with proteins allowing conversion from transcriptional initiation to elongation.

Figure 5 eRNAs. (A) ChIP-Seq analysis identifies eRNA producing enhancers by enrichment for H3K27Ac and RNA pol II. (B) Model of eRNA function at Nanog function. eRNAs appear to facilitate or stabilize chromatin loops between specific enhancer:gene promoter combinations. Teaching Point: eRNAs can be identified through ChIP-Seq analysis and may play a role in regulation gene expression changes.

Figure 6 Proposed models of alteration of enhancer function leads to cancer development. (A) Activation of enhancers results in the opening of chromatin, leading to the greater likelihood of chromosomal rearrangements that ultimately lead to aneuploidy (21). (B) Duplication of enhancer sequences that regulate MYC leads to increased MYC expression that drives epithelial cancers (121). (C) Translocation of G2DHE (a GATA2 enhancer) near the EVI1 locus leads to overexpression of the EVI1 oncogene, promoting myeloid and B-cell leukemic development (119). (D) Heterozygous loss of cohesin leads to changes in nuclear architecture that can alter enhancer-promoter interactions and lead to gene expression changes that promote myeloid transformation. Teaching point: There are many routes that enhancer perturbation can lead to cancer development.

 


Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Puja Agrawal, Katelyn E. Heimbruch, Sridhar Rao. Genome‐Wide Maps of Transcription Regulatory Elements and Transcription Enhancers in Development and Disease. Compr Physiol 2018, 9: 439-455. doi: 10.1002/cphy.c180028