Browse > Article
http://dx.doi.org/10.14348/molcells.2018.2297

Targeting Super-Enhancers for Disease Treatment and Diagnosis  

Shin, Ha Youn (Department of Biomedical Science and Engineering, Konkuk University)
Abstract
The transcriptional regulation of genes determines the fate of animal cell differentiation and subsequent organ development. With the recent progress in genome-wide technologies, the genomic landscapes of enhancers have been broadly explored in mammalian genomes, which led to the discovery of novel specific subsets of enhancers, termed super-enhancers. Super-enhancers are large clusters of enhancers covering the long region of regulatory DNA and are densely occupied by transcription factors, active histone marks, and co-activators. Accumulating evidence points to the critical role that super-enhancers play in cell type-specific development and differentiation, as well as in the development of various diseases. Here, I provide a comprehensive description of the optimal approach for identifying functional units of super-enhancers and their unique chromatin features in normal development and in diseases, including cancers. I also review the recent updated knowledge on novel approaches of targeting super-enhancers for the treatment of specific diseases, such as small-molecule inhibitors and potential gene therapy. This review will provide perspectives on using super-enhancers as biomarkers to develop novel disease diagnostic tools and establish new directions in clinical therapeutic strategies.
Keywords
cell identity; clinical therapeutics; diseases; disease diagnosis; super-enhancer;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Wong, R.W.J., Ngoc, P.C.T., Leong, W.Z., Yam, A.W.Y., Zhang, T., Asamitsu, K., Iida, S., Okamoto, T., Ueda, R., Gray, N.S., et al. (2017). Enhancer profiling identifies critical cancer genes and characterizes cell identity in adult T-cell leukemia. Blood 130, 2326-2338   DOI
2 Wyce, A., Ganji, G., Smitheman, K.N., Chung, C.W., Korenchuk, S., Bai, Y., Barbash, O., Le, B., Craggs, P.D., McCabe, M.T., et al. (2013). BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models. PLoS One 8, e72967.   DOI
3 Yin, J.W., and Wang, G. (2014). The Mediator complex: a master coordinator of transcription and cell lineage development. Development 141, 977-987.   DOI
4 Yokoyama, Y., Zhu, H., Lee, J.H., Kossenkov, A.V., Wu, S.Y., Wickramasinghe, J.M., Yin, X., Palozola, K.C., Gardini, A., Showe, L.C., et al. (2016). BET inhibitors suppress ALDH activity by targeting ALDH1A1 super-enhancer in ovarian cancer. Cancer Res 76, 6320-6330.   DOI
5 Zeng, L., and Zhou, M.M. (2002). Bromodomain: an acetyl-lysine binding domain. FEBS Lett. 513, 124-128.   DOI
6 Zhang, X., Choi, P.S., Francis, J.M., Imielinski, M., Watanabe, H., Cherniack, A.D., and Meyerson, M. (2016). Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers. Nat. Genet. 48, 176-182.   DOI
7 Ko, J.Y., Oh, S., and Yoo, K.H. (2017). Functional Enhancers As Master Regulators of Tissue-Specific Gene Regulation and Cancer Development. Mol. Cells 40, 169-177.
8 Kulaeva, O.I., Nizovtseva, E.V., Polikanov, Y.S., Ulianov, S.V. and Studitsky, V.M. (2012). Distant activation of transcription: mechanisms of enhancer action. Mol. Cell Biol. 32, 4892-4897.   DOI
9 Kwiatkowski, N., Zhang, T., Rahl, P.B., Abraham, B.J., Reddy, J., Ficarro, S.B., Dastur, A., Amzallag, A., Ramaswamy, S., Tesar, B., et al. (2014). Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 511, 616-620.   DOI
10 Le Gras, S., Keime, C., Anthony, A., Lotz, C., De Longprez, L., Brouillet, E., Cassel, J.C., Boutillier, A.L. and Merienne, K. (2017). Altered enhancer transcription underlies Huntington's disease striatal transcriptional signature. Sci. Rep. 7, 42875.   DOI
11 Li, Y., Rivera, C.M., Ishii, H., Jin, F., Selvaraj, S., Lee, A.Y., Dixon, J.R., and Ren, B. (2014). CRISPR reveals a distal super-enhancer required for Sox2 expression in mouse embryonic stem cells. PLoS One 9, e114485.   DOI
12 Achour, M., Le Gras, S., Keime, C., Parmentier, F., Lejeune, F.X., Boutillier, A.L., Neri, C., Davidson, I., and Merienne, K. (2015). Neuronal identity genes regulated by super-enhancers are preferentially down-regulated in the striatum of Huntington's disease mice. Hum. Mol. Genet. 24, 3481-3496.   DOI
13 Adam, R.C., Yang, H., Rockowitz, S., Larsen, S.B., Nikolova, M., Oristian, D.S., Polak, L., Kadaja, M., Asare, A., Zheng, D., et al. (2015). Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice. Nature 521, 366-370.   DOI
14 Lee, H.K., Willi, M., Wang, C., Yang, C.M., Smith, H.E., Liu, C., and Hennighausen, L. (2017). Functional assessment of CTCF sites at cytokine-sensing mammary enhancers using CRISPR/Cas9 gene editing in mice. Nucleic Acids Res. 45, 4606-4618.   DOI
15 Levine, M. (2010). Transcriptional enhancers in animal development and evolution. Curr. Biol. 20, R754-763.   DOI
16 Levine, M., Cattoglio, C., and Tjian, R. (2014). Looping back to leap forward: transcription enters a new era. Cell 157, 13-25.   DOI
17 Liang, J., Zhou, H., Gerdt, C., Tan, M., Colson, T., Kaye, K.M., Kieff, E., and Zhao, B. (2016). Epstein-Barr virus super-enhancer eRNAs are essential for MYC oncogene expression and lymphoblast proliferation. Proc. Natl. Acad. Sci. USA 113, 14121-14126.   DOI
18 Amorim, S., Stathis, A., Gleeson, M., Iyengar, S., Magarotto, V., Leleu, X., Morschhauser, F., Karlin, L., Broussais, F., Rezai, K., et al. (2016). Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol. 3, e196-204.   DOI
19 Loven, J., Hoke, H.A., Lin, C.Y., Lau, A., Orlando, D.A., Vakoc, C.R., Bradner, J.E., Lee, T.I., and Young, R.A. (2013). Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320-334.   DOI
20 Mansour, M.R., Abraham, B.J., Anders, L., Berezovskaya, A., Gutierrez, A., Durbin, A.D., Etchin, J., Lawton, L., Sallan, S.E., Silverman, L.B., et al. (2014). Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346, 1373-1377.   DOI
21 Banerji, J., Rusconi, S., and Schaffner, W. (1981). Expression of a beta-globin gene is enhanced by remote SV40 DNA sequences. Cell 27, 299-308.   DOI
22 Ong, C.T., and Corces, V.G. (2012). Enhancers: emerging roles in cell fate specification. EMBO Rep. 13, 423-430.   DOI
23 Northcott, P.A., Lee, C., Zichner, T., Stutz, A.M., Erkek, S., Kawauchi, D., Shih, D.J., Hovestadt, V., Zapatka, M., Sturm, D., et al. (2014). Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511, 428-434.   DOI
24 Oldridge, D.A., Wood, A.C., Weichert-Leahey, N., Crimmins, I., Sussman, R., Winter, C., McDaniel, L.D., Diamond, M., Hart, L.S., Zhu, S., et al. (2015). Genetic predisposition to neuroblastoma mediated by a LMO1 super-enhancer polymorphism. Nature 528, 418-421.   DOI
25 Ong, C.T., and Corces, V.G. (2011). Enhancer function: new insights into the regulation of tissue-specific gene expression. Nat. Rev. Genet. 12, 283-293.
26 Parker, S.C., Stitzel, M.L., Taylor, D.L., Orozco, J.M., Erdos, M.R., Akiyama, J.A., van Bueren, K.L., Chines, P.S., Narisu, N., Program, N.C.S., et al. (2013). Chromatin stretch enhancer states drive cell-specific gene regulation and harbor human disease risk variants. Proc. Natl. Acad. Sci. USA 110, 17921-17926.   DOI
27 Chipumuro, E., Marco, E., Christensen, C.L., Kwiatkowski, N., Zhang, T., Hatheway, C.M., Abraham, B.J., Sharma, B., Yeung, C., Altabef, A., et al. (2014). CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 159, 1126-1139.   DOI
28 Banerji, J., Olson, L., and Schaffner, W. (1983). A lymphocyte-specific cellular enhancer is located downstream of the joining region in immunoglobulin heavy chain genes. Cell 33, 729-740.   DOI
29 Carter, D., Chakalova, L., Osborne, C.S., Dai, Y.F., and Fraser, P. (2002). Long-range chromatin regulatory interactions in vivo. Nat. Genet. 32, 623-626.   DOI
30 Cavalli, G., Hayashi, M., Jin, Y., Yorgov, D., Santorico, S.A., Holcomb, C., Rastrou, M., Erlich, H., Tengesdal, I.W., Dagna, L., et al. (2016). MHC class II super-enhancer increases surface expression of HLA-DR and HLA-DQ and affects cytokine production in autoimmune vitiligo. Proc. Natl. Acad. Sci. USA 113, 1363-1368.   DOI
31 Christensen, C.L., Kwiatkowski, N., Abraham, B.J., Carretero, J., Al-Shahrour, F., Zhang, T., Chipumuro, E., Herter-Sprie, G.S., Akbay, E.A., Altabef, A., et al. (2014). Targeting transcriptional addictions in small cell lung cancer with a covalent CDK7 inhibitor. Cancer Cell 26, 909-922.   DOI
32 Pott, S. and Lieb, J.D. (2015). What are super-enhancers? Nat Genet 47, 8-12.   DOI
33 Peeters, J.G., Vervoort, S.J., Tan, S.C., Mijnheer, G., de Roock, S., Vastert, S.J., Nieuwenhuis, E.E., van Wijk, F., Prakken, B.J., Creyghton, M.P., et al. (2015). Inhibition of super-enhancer activity in autoinflammatory site-derived T cells reduces disease-associated gene expression. Cell Rep. 12, 1986-1996.   DOI
34 Pefanis, E., Wang, J., Rothschild, G., Lim, J., Kazadi, D., Sun, J., Federation, A., Chao, J., Elliott, O., Liu, Z.P., et al. (2015). RNA exosome-regulated long non-coding RNA transcription controls super-enhancer activity. Cell 161, 774-789.   DOI
35 Pelish, H.E., Liau, B.B., Nitulescu, II, Tangpeerachaikul, A., Poss, Z.C., Da Silva, D.H., Caruso, B.T., Arefolov, A., Fadeyi, O., Christie, A.L., et al. (2015). Mediator kinase inhibition further activates super-enhancer-associated genes in AML. Nature 526, 273-276.   DOI
36 Sengupta, S., and George, R.E. (2017). Super-enhancer-driven transcriptional dependencies in cancer. Trends Cancer 3, 269-281.   DOI
37 Sengupta, D., Kannan, A., Kern, M., Moreno, M.A., Vural, E., Stack, B., Jr., Suen, J.Y., Tackett, A.J. and Gao, L. (2015). Disruption of BRD4 at H3K27Ac-enriched enhancer region correlates with decreased c-Myc expression in Merkel cell carcinoma. Epigenetics 10, 460-466.   DOI
38 Shi, J., Whyte, W.A., Zepeda-Mendoza, C.J., Milazzo, J.P., Shen, C., Roe, J.S., Minder, J.L., Mercan, F., Wang, E., Eckersley-Maslin, M.A., et al. (2013). Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation. Genes Dev 27, 2648-2662.   DOI
39 Shin, H.Y., Willi, M., Yoo, K.H., Zeng, X., Wang, C., Metser, G., and Hennighausen, L. (2016). Hierarchy within the mammary STAT5-driven Wap super-enhancer. Nat Genet 48, 904-911.   DOI
40 Dowen, J.M., Fan, Z.P., Hnisz, D., Ren, G., Abraham, B.J., Zhang, L.N., Weintraub, A.S., Schujiers, J., Lee, T.I., Zhao, K., et al. (2014). Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell 159, 374-387.   DOI
41 Heinz, S., Romanoski, C.E., Benner, C., and Glass, C.K. (2015). The selection and function of cell type-specific enhancers. Nat. Rev. Mol. Cell Biol. 16, 144-154.   DOI
42 Drier, Y., Cotton, M.J., Williamson, K.E., Gillespie, S.M., Ryan, R.J., Kluk, M.J., Carey, C.D., Rodig, S.J., Sholl, L.M., Afrogheh, A.H., et al. (2016). An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma. Nat. Genet. 48, 265-272.   DOI
43 Groschel, S., Sanders, M.A., Hoogenboezem, R., de Wit, E., Bouwman, B.A.M., Erpelinck, C., van der Velden, V.H.J., Havermans, M., Avellino, R., van Lom, K., et al. (2014). A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell 157, 369-381.   DOI
44 Hay, D., Hughes, J.R., Babbs, C., Davies, J.O.J., Graham, B.J., Hanssen, L., Kassouf, M.T., Marieke Oudelaar, A.M., Sharpe, J.A., Suciu, M.C., et al. (2016). Genetic dissection of the alpha-globin super-enhancer in vivo. Nat. Genet. 48, 895-903.   DOI
45 Henssen, A., Althoff, K., Odersky, A., Beckers, A., Koche, R., Speleman, F., Schafers, S., Bell, E., Nortmeyer, M., Westermann, F., et al. (2016). Targeting MYCN-driven transcription by BETbromodomain inhibition. Clin. Cancer Res. 22, 2470-2481.   DOI
46 Herz, H.M., Hu, D., and Shilatifard, A. (2014). Enhancer malfunction in cancer. Mol. Cell 53, 859-866.   DOI
47 Hnisz, D., Abraham, B.J., Lee, T.I., Lau, A., Saint-Andre, V., Sigova, A.A., Hoke, H.A., and Young, R.A. (2013). Super-enhancers in the control of cell identity and disease. Cell 155, 934-947.   DOI
48 Tolhuis, B., Palstra, R.J., Splinter, E., Grosveld, F., and de Laat, W. (2002). Looping and interaction between hypersensitive sites in the active beta-globin locus. Mol. Cell 10, 1453-1465.   DOI
49 Shlyueva, D., Stampfel, G., and Stark, A. (2014). Transcriptional enhancers: from properties to genome-wide predictions. Nat. Rev. Genet. 15, 272-286.   DOI
50 Siersbaek, R., Rabiee, A., Nielsen, R., Sidoli, S., Traynor, S., Loft, A., La Cour Poulsen, L., Rogowska-Wrzesinska, A., Jensen, O.N., and Mandrup, S. (2014). Transcription factor cooperativity in early adipogenic hotspots and super-enhancers. Cell Rep. 7, 1443-1455.   DOI
51 Vahedi, G., Kanno, Y., Furumoto, Y., Jiang, K., Parker, S.C., Erdos, M.R., Davis, S.R., Roychoudhuri, R., Restifo, N.P., Gadina, M., et al. (2015). Super-enhancers delineate disease-associated regulatory nodes in T cells. Nature 520, 558-562.   DOI
52 Walker, B.A., Wardell, C.P., Brioli, A., Boyle, E., Kaiser, M.F., Begum, D.B., Dahir, N.B., Johnson, D.C., Ross, F.M., Davies, F.E., et al. (2014). Translocations at 8q24 juxtapose MYC with genes that harbor superenhancers resulting in overexpression and poor prognosis in myeloma patients. Blood Cancer J 4, e191.   DOI
53 Whyte, W.A., Orlando, D.A., Hnisz, D., Abraham, B.J., Lin, C.Y., Kagey, M.H., Rahl, P.B., Lee, T.I., and Young, R.A. (2013). Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307-319.   DOI
54 Willi, M., Yoo, K.H., Reinisch, F., Kuhns, T.M., Lee, H.K., Wang, C., and Hennighausen, L. (2017). Facultative CTCF sites moderate mammary super-enhancer activity and regulate juxtaposed gene in non-mammary cells. Nat. Commun. 8, 16069.   DOI
55 Kagey, M.H., Newman, J.J., Bilodeau, S., Zhan, Y., Orlando, D.A., van Berkum, N.L., Ebmeier, C.C., Goossens, J., Rahl, P.B., Levine, S.S., et al. (2010). Mediator and cohesin connect gene expression and chromatin architecture. Nature 467, 430-435.   DOI
56 Huang, J., Liu, X., Li, D., Shao, Z., Cao, H., Zhang, Y., Trompouki, E., Bowman, T.V., Zon, L.I., Yuan, G.C., et al. (2016). Dynamic control of enhancer repertoires drives lineage and stage-specific transcription during hematopoiesis. Dev. Cell 36, 9-23.   DOI
57 Jiang, Y.Y., Lin, D.C., Mayakonda, A., Hazawa, M., Ding, L.W., Chien, W.W., Xu, L., Chen, Y., Xiao, J.F., Senapedis, W., et al. (2017). Targeting super-enhancer-associated oncogenes in oesophageal squamous cell carcinoma. Gut 66, 1358-1368.   DOI
58 Jiao, W., Chen, Y., Song, H., Li, D., Mei, H., Yang, F., Fang, E., Wang, X., Huang, K., Zheng, L., et al. (2018). HPSE enhancer RNA promotes cancer progression through driving chromatin looping and regulating hnRNPU/p300/EGR1/HPSE axis. Oncogene [Epub ahead of print].
59 Katerndahl, C.D.S., Heltemes-Harris, L.M., Willette, M.J.L., Henzler, C.M., Frietze, S., Yang, R., Schjerven, H., Silverstein, K.A.T., Ramsey, L.B., Hubbard, G., et al. (2017). Antagonism of B cell enhancer networks by STAT5 drives leukemia and poor patient survival. Nat Immunol 18, 694-704.   DOI
60 Kennedy, A.L., Vallurupalli, M., Chen, L., Crompton, B., Cowley, G., Vazquez, F., Weir, B.A., Tsherniak, A., Parasuraman, S., Kim, S., et al. (2015). Functional, chemical genomic, and super-enhancer screening identify sensitivity to cyclin D1/CDK4 pathway inhibition in Ewing sarcoma. Oncotarget 6, 30178-30193.
61 Kim, T.K., Hemberg, M., Gray, J.M., Costa, A.M., Bear, D.M., Wu, J., Harmin, D.A., Laptewicz, M., Barbara-Haley, K., Kuersten, S., et al. (2010). Widespread transcription at neuronal activity-regulated enhancers. Nature 465, 182-187.   DOI