83
Views
12
CrossRef citations to date
0
Altmetric
Research Article

Loss of Estrogen-Related Receptor Alpha Facilitates Angiogenesis in Endothelial Cells

, , , , , , , , & show all
Article: e00411-18 | Received 16 Aug 2018, Accepted 11 Dec 2018, Published online: 03 Mar 2023

REFERENCES

  • Rodrigues SF, Granger DN. 2015. Blood cells and endothelial barrier function. Tissue Barriers 3:e978720. https://doi.org/10.4161/21688370.2014.978720.
  • Förstermann U, Sessa WC. 2012. Nitric oxide synthases: regulation and function. Eur Heart J 33:829–837. https://doi.org/10.1093/eurheartj/ehr304.
  • Pober JS, Sessa WC. 2007. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol 7:803–815. https://doi.org/10.1038/nri2171.
  • Risau W. 1997. Mechanisms of angiogenesis. Nature 386:671–674. https://doi.org/10.1038/386671a0.
  • Blanco R, Gerhardt H. 2013. VEGF and Notch in tip and stalk cell selection. Cold Spring Harb Perspect Med 3:a006569. https://doi.org/10.1101/cshperspect.a006569.
  • Marcelo KL, Goldie LC, Hirschi KK. 2013. Regulation of endothelial cell differentiation and specification. Circ Res 112:1272–1287. https://doi.org/10.1161/CIRCRESAHA.113.300506.
  • Patel-Hett S, Damore PA. 2011. Signal transduction in vasculogenesis and developmental angiogenesis. Int J Dev Biol 55:353–363. https://doi.org/10.1387/ijdb.103213sp.
  • Ucuzian AA, Gassman AA, East AT, Greisler HP. 2010. Molecular mediators of angiogenesis. J Burn Care Res 31:158–175. https://doi.org/10.1097/BCR.0b013e3181c7ed82.
  • Potente M, Gerhardt H, Carmeliet P. 2011. Basic and therapeutic aspects of angiogenesis. Cell 146:873–887. https://doi.org/10.1016/j.cell.2011.08.039.
  • Jakobsson L, Bentley K, Gerhardt H. 2009. VEGFRs and Notch: a dynamic collaboration in vascular patterning. Biochem Soc Trans 37:1233–1236. https://doi.org/10.1042/BST0371233.
  • Park C, Kim TM, Malik AB. 2013. Transcriptional regulation of endothelial cell and vascular development. Circ Res 112:1380–1400. https://doi.org/10.1161/CIRCRESAHA.113.301078.
  • De Val S, Black BL. 2009. Transcriptional control of endothelial cell development. Dev Cell 16:180–195. https://doi.org/10.1016/j.devcel.2009.01.014.
  • Bautch VL. 2012. VEGF-directed blood vessel patterning: from cells to organism. Cold Spring Harb Perspect Med 2:a006452. https://doi.org/10.1101/cshperspect.a006452.
  • Jeltsch M, Leppanen VM, Saharinen P, Alitalo K. 2013. Receptor tyrosine kinase-mediated angiogenesis. Cold Spring Harb Perspect Biol 5:a009183. https://doi.org/10.1101/cshperspect.a009183.
  • Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RM. 1995. The nuclear receptor superfamily: the second decade. Cell 83:835–839. https://doi.org/10.1016/0092-8674(95)90199-X.
  • Suzuki T, Nakamura Y, Moriya T, Sasano H. 2003. Effects of steroid hormones on vascular functions. Microsc Res Tech 60:76–84. https://doi.org/10.1002/jemt.10246.
  • Brouchet L, Krust A, Dupont S, Chambon P, Bayard F, Arnal JF. 2001. Estradiol accelerates reendothelialization in mouse carotid artery through estrogen receptor-alpha but not estrogen receptor-beta. Circulation 103:423–428. https://doi.org/10.1161/01.CIR.103.3.423.
  • Caulin-Glaser T, Farrell WJ, Pfau SE, Zaret B, Bunger K, Setaro JF, Brennan JJ, Bender JR, Cleman MW, Cabin HS, Remetz MS. 1998. Modulation of circulating cellular adhesion molecules in postmenopausal women with coronary artery disease. J Am Coll Cardiol 31:1555–1560. https://doi.org/10.1016/S0735-1097(98)00145-4.
  • Jun SS, Chen Z, Pace MC, Shaul PW. 1998. Estrogen upregulates cyclooxygenase-1 gene expression in ovine fetal pulmonary artery endothelium. J Clin Investig 102:176–183. https://doi.org/10.1172/JCI2034.
  • Losordo DW, Isner JM. 2001. Estrogen and angiogenesis: a review. Arterioscler Thromb Vasc Biol 21:6–12. https://doi.org/10.1161/01.ATV.21.1.6.
  • Morales DE, McGowan KA, Grant DS, Maheshwari S, Bhartiya D, Cid MC, Kleinman HK, Schnaper HW. 1995. Estrogen promotes angiogenic activity in human umbilical vein endothelial cells in vitro and in a murine model. Circulation 91:755–763. https://doi.org/10.1161/01.CIR.91.3.755.
  • Prabhushankar R, Krueger C, Manrique C. 2014. Membrane estrogen receptors: their role in blood pressure regulation and cardiovascular disease. Curr Hypertens Rep 16:013–0408.
  • Ingegno MD, Money SR, Thelmo W, Greene GL, Davidian M, Jaffe BM, Pertschuk LP. 1988. Progesterone receptors in the human heart and great vessels. Lab Investig 59:353–356.
  • Schnaper HW, McGuire J, Runyan C, Hubchak SC. 2000. Sex steroids and the endothelium. Curr Med Chem 7:519–531. https://doi.org/10.2174/0929867003374949.
  • Lee WS, Liu CW, Juan SH, Liang YC, Ho PY, Lee YH. 2003. Molecular mechanism of progesterone-induced antiproliferation in rat aortic smooth muscle cells. Endocrinology 144:2785–2790. https://doi.org/10.1210/en.2003-0045.
  • Wu J, Hadoke PWF, Takov K, Korczak A, Denvir MA, Smith LB. 2016. Influence of androgen receptor in vascular cells on reperfusion following hindlimb ischaemia. PLoS One 11:e0154987. https://doi.org/10.1371/journal.pone.0154987.
  • Takov K, Wu J, Denvir MA, Smith LB, Hadoke PWF. 2018. The role of androgen receptors in atherosclerosis. Mol Cell Endocrinol 465:82–91. https://doi.org/10.1016/j.mce.2017.10.006.
  • Ullian ME. 1999. The role of corticosteriods in the regulation of vascular tone. Cardiovasc Res 41:55–64. https://doi.org/10.1016/S0008-6363(98)00230-2.
  • Logie JJ, Ali S, Marshall KM, Heck MMS, Walker BR, Hadoke PWF. 2010. Glucocorticoid-mediated inhibition of angiogenic changes in human endothelial cells is not caused by reductions in cell proliferation or migration. PLoS One 5:e14476. https://doi.org/10.1371/journal.pone.0014476.
  • Yano A, Fujii Y, Iwai A, Kageyama Y, Kihara K. 2006. Glucocorticoids suppress tumor angiogenesis and in vivo growth of prostate cancer cells. Clin Cancer Res 12:3003–3009. https://doi.org/10.1158/1078-0432.CCR-05-2085.
  • Lother A, Moser M, Bode C, Feldman RD, Hein L. 2015. Mineralocorticoids in the heart and vasculature: new insights for old hormones. Annu Rev Pharmacol Toxicol 55:289–312. https://doi.org/10.1146/annurev-pharmtox-010814-124302.
  • Giguere V. 1999. Orphan nuclear receptors: from gene to function. Endocr Rev 20:689–725. https://doi.org/10.1210/er.20.5.689.
  • Mullican SE, DiSpirito JR, Lazar MA. 2013. The orphan nuclear receptors at their 25th year reunion. J Mol Endocrinol 51:T115–T140. https://doi.org/10.1530/JME-13-0212.
  • Kotlinowski J, Jozkowicz A. 2016. PPAR gamma and angiogenesis: endothelial cells perspective. J Diabetes Res 2016:8492353. https://doi.org/10.1155/2016/8492353.
  • Hong HK, Cho YM, Park KH, Lee CT, Lee HK, Park KS. 2003. Peroxisome proliferator-activated receptor gamma mediated inhibition of plasminogen activator inhibitor type 1 production and proliferation of human umbilical vein endothelial cells. Diabetes Res Clin Pract 62:1–8. https://doi.org/10.1016/S0168-8227(03)00142-6.
  • Zhao Y, Bruemmer D. 2010. NR4A orphan nuclear receptors: transcriptional regulators of gene expression in metabolism and vascular biology. Arterioscler Thromb Vasc Biol 30:1535–1541. https://doi.org/10.1161/ATVBAHA.109.191163.
  • Pereira FA, Qiu Y, Zhou G, Tsai MJ, Tsai SY. 1999. The orphan nuclear receptor COUP-TFII is required for angiogenesis and heart development. Genes Dev 13:1037–1049. https://doi.org/10.1101/gad.13.8.1037.
  • Giguere V. 2008. Transcriptional control of energy homeostasis by the estrogen-related receptors. Endocr Rev 29:677–696. https://doi.org/10.1210/er.2008-0017.
  • Giguere V, Yang N, Segui P, Evans RM. 1988. Identification of a new class of steroid hormone receptors. Nature 331:91–94. https://doi.org/10.1038/331091a0.
  • Dufour CR, Wilson BJ, Huss JM, Kelly DP, Alaynick WA, Downes M, Evans RM, Blanchette M, Giguere V. 2007. Genome-wide orchestration of cardiac functions by the orphan nuclear receptors ERRalpha and gamma. Cell Metab 5:345–356. https://doi.org/10.1016/j.cmet.2007.03.007.
  • Audet-Walsh É, Giguére V. 2015. The multiple universes of estrogen-related receptor alpha and gamma in metabolic control and related diseases. Acta Pharmacol Sin 36:51–61. https://doi.org/10.1038/aps.2014.121.
  • Huss JM, Kopp RP, Kelly DP. 2002. Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identification of novel leucine-rich interaction motif within PGC-1alpha. J Biol Chem 277:40265–40274. https://doi.org/10.1074/jbc.M206324200.
  • Kamei Y, Ohizumi H, Fujitani Y, Nemoto T, Tanaka T, Takahashi N, Kawada T, Miyoshi M, Ezaki O, Kakizuka A. 2003. PPARgamma coactivator 1beta/ERR ligand 1 is an ERR protein ligand, whose expression induces a high-energy expenditure and antagonizes obesity. Proc Natl Acad Sci U S A 100:12378–12383. https://doi.org/10.1073/pnas.2135217100.
  • Schreiber SN, Emter R, Hock MB, Knutti D, Cardenas J, Podvinec M, Oakeley EJ, Kralli A. 2004. The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis. Proc Natl Acad Sci U S A 101:6472–6477. https://doi.org/10.1073/pnas.0308686101.
  • Perry MC, Dufour CR, Tam IS, B'Chir W, Giguere V. 2014. Estrogen-related receptor-alpha coordinates transcriptional programs essential for exercise tolerance and muscle fitness. Mol Endocrinol 28:2060–2071. https://doi.org/10.1210/me.2014-1281.
  • Narkar VA, Fan W, Downes M, Yu RT, Jonker JW, Alaynick WA, Banayo E, Karunasiri MS, Lorca S, Evans RM. 2011. Exercise and PGC-1alpha-independent synchronization of type I muscle metabolism and vasculature by ERRgamma. Cell Metab 13:283–293. https://doi.org/10.1016/j.cmet.2011.01.019.
  • Luo J, Sladek R, Carrier J, Bader JA, Richard D, Giguere V. 2003. Reduced fat mass in mice lacking orphan nuclear receptor estrogen-related receptor alpha. Mol Cell Biol 23:7947–7956. https://doi.org/10.1128/MCB.23.22.7947-7956.2003.
  • Powelka AM, Seth A, Virbasius JV, Kiskinis E, Nicoloro SM, Guilherme A, Tang X, Straubhaar J, Cherniack AD, Parker MG, Czech MP. 2006. Suppression of oxidative metabolism and mitochondrial biogenesis by the transcriptional corepressor RIP140 in mouse adipocytes. J Clin Investig 116:125–136.
  • Villena JA, Hock MB, Chang WY, Barcas JE, Giguere V, Kralli A. 2007. Orphan nuclear receptor estrogen-related receptor alpha is essential for adaptive thermogenesis. Proc Natl Acad Sci U S A 104:1418–1423. https://doi.org/10.1073/pnas.0607696104.
  • Huss JM, Garbacz WG, Xie W. 2015. Constitutive activities of estrogen-related receptors: transcriptional regulation of metabolism by the ERR pathways in health and disease. Biochim Biophys Acta 9:24.
  • Sonoda J, Laganiere J, Mehl IR, Barish GD, Chong LW, Li X, Scheffler IE, Mock DC, Bataille AR, Robert F, Lee CH, Giguere V, Evans RM. 2007. Nuclear receptor ERR alpha and coactivator PGC-1 beta are effectors of IFN-gamma-induced host defense. Genes Dev 21:1909–1920. https://doi.org/10.1101/gad.1553007.
  • Michalek RD, Gerriets VA, Nichols AG, Inoue M, Kazmin D, Chang CY, Dwyer MA, Nelson ER, Pollizzi KN, Ilkayeva O, Giguere V, Zuercher WJ, Powell JD, Shinohara ML, McDonnell DP, Rathmell JC. 2011. Estrogen-related receptor-alpha is a metabolic regulator of effector T-cell activation and differentiation. Proc Natl Acad Sci U S A 108:18348–18353. https://doi.org/10.1073/pnas.1108856108.
  • Matsakas A, Yadav V, Lorca S, Evans RM, Narkar VA. 2012. Revascularization of ischemic skeletal muscle by estrogen-related receptor-gamma. Circ Res 110:1087–1096. https://doi.org/10.1161/CIRCRESAHA.112.266478.
  • Stein RA, Gaillard S, McDonnell DP. 2009. Estrogen-related receptor alpha induces the expression of vascular endothelial growth factor in breast cancer cells. J Steroid Biochem Mol Biol 114:106–112. https://doi.org/10.1016/j.jsbmb.2009.02.010.
  • Liang J, Han F, Chen Y. 2013. Transcriptional regulation of VEGF expression by estrogen-related receptor γ. Acta Pharmaceutica Sinica B 3:373–380. https://doi.org/10.1016/j.apsb.2013.09.001.
  • Sobczak M, Dargatz J, Chrzanowska-Wodnicka M. 2010. Isolation and culture of pulmonary endothelial cells from neonatal mice. J Vis Exp 14:2316.
  • Korff T, Augustin HG. 1999. Tensional forces in fibrillar extracellular matrices control directional capillary sprouting. J Cell Sci 112:3249–3258.
  • Pfisterer L, Korff T. 2016. Spheroid-based in vitro angiogenesis model, p 167–177. In Martin SG, Hewett PW (ed), Angiogenesis protocols. Springer New York, New York, NY. https://doi.org/10.1007/978-1-4939-3628-1_11.
  • Rangwala SM, Wang X, Calvo JA, Lindsley L, Zhang Y, Deyneko G, Beaulieu V, Gao J, Turner G, Markovits J. 2010. Estrogen-related receptor gamma is a key regulator of muscle mitochondrial activity and oxidative capacity. J Biol Chem 285:22619–22629. https://doi.org/10.1074/jbc.M110.125401.
  • Alaynick WA, Kondo RP, Xie W, He W, Dufour CR, Downes M, Jonker JW, Giles W, Naviaux RK, Giguere V, Evans RM. 2007. ERRgamma directs and maintains the transition to oxidative metabolism in the postnatal heart. Cell Metab 6:13–24. https://doi.org/10.1016/j.cmet.2007.06.007.
  • Vega RB, Kelly DP. 1997. A role for estrogen-related receptor alpha in the control of mitochondrial fatty acid beta-oxidation during brown adipocyte differentiation. J Biol Chem 272:31693–31699. https://doi.org/10.1074/jbc.272.50.31693.
  • Cruys B, Wong BW, Kuchnio A, Verdegem D, Cantelmo AR, Conradi LC, Vandekeere S, Bouche A, Cornelissen I, Vinckier S, Merks RM, Dejana E, Gerhardt H, Dewerchin M, Bentley K, Carmeliet P. 2016. Glycolytic regulation of cell rearrangement in angiogenesis. Nat Commun 7:12240. https://doi.org/10.1038/ncomms12240.
  • De Bock K, Georgiadou M, Carmeliet P. 2013. Role of endothelial cell metabolism in vessel sprouting. Cell Metab 18:634–647. https://doi.org/10.1016/j.cmet.2013.08.001.
  • Treps L, Conradi LC, Harjes U, Carmeliet P. 2016. Manipulating angiogenesis by targeting endothelial metabolism: hitting the engine rather than the drivers-a new perspective? Pharmacol Rev 68:872–887. https://doi.org/10.1124/pr.116.012492.
  • Rothhammer T, Bataille F, Spruss T, Eissner G, Bosserhoff AK. 2007. Functional implication of BMP4 expression on angiogenesis in malignant melanoma. Oncogene 26:4158–4170. https://doi.org/10.1038/sj.onc.1210182.
  • Yadav SS, Narayan G. 2014. Role of ROBO4 signalling in developmental and pathological angiogenesis. Biomed Res Int 2014:683025. https://doi.org/10.1155/2014/683025.
  • Zhou Y, Williams J, Smallwood PM, Nathans J. 2015. Sox7, Sox17, and Sox18 cooperatively regulate vascular development in the mouse retina. PLoS One 10:e0143650. https://doi.org/10.1371/journal.pone.0143650.
  • Fukumura D, Gohongi T, Kadambi A, Izumi Y, Ang J, Yun CO, Buerk DG, Huang PL, Jain RK. 2001. Predominant role of endothelial nitric oxide synthase in vascular endothelial growth factor-induced angiogenesis and vascular permeability. Proc Natl Acad Sci U S A 98:2604–2609. https://doi.org/10.1073/pnas.041359198.
  • Lamalice L, Le Boeuf F, Huot J. 2007. Endothelial cell migration during angiogenesis. Circ Res 100:782–794. https://doi.org/10.1161/01.RES.0000259593.07661.1e.
  • Cao G, O'Brien CD, Zhou Z, Sanders SM, Greenbaum JN, Makrigiannakis A, DeLisser HM. 2002. Involvement of human PECAM-1 in angiogenesis and in vitro endothelial cell migration. Am J Physiol Cell Physiol 282:C1181–C1190. https://doi.org/10.1152/ajpcell.00524.2001.
  • van Hinsbergh VW, Koolwijk P. 2008. Endothelial sprouting and angiogenesis: matrix metalloproteinases in the lead. Cardiovasc Res 78:203–212. https://doi.org/10.1093/cvr/cvm102.
  • Ishida T, Kundu RK, Yang E, Hirata K, Ho YD, Quertermous T. 2003. Targeted disruption of endothelial cell-selective adhesion molecule inhibits angiogenic processes in vitro and in vivo. J Biol Chem 278:34598–34604. https://doi.org/10.1074/jbc.M304890200.
  • Yang H, Zhang H, Ge S, Ning T, Bai M, Li J, Li S, Sun W, Deng T, Zhang L, Ying G, Ba Y. 2018. Exosome-derived miR-130a activates angiogenesis in gastric cancer by targeting C-MYB in vascular endothelial cells. Mol Ther 26:2466–2475. https://doi.org/10.1016/j.ymthe.2018.07.023.
  • ENCODE Project Consortium. 2012. An integrated encyclopedia of DNA elements in the human genome. Nature 489:57–74. https://doi.org/10.1038/nature11247.
  • Audet-Walsh É, Papadopoli DJ, Gravel S-P, Yee T, Bridon G, Caron M, Bourque G, Giguère V, St-Pierre J. 2016. The PGC-1alpha/ERRalpha axis represses one-carbon metabolism and promotes sensitivity to anti-folate therapy in breast cancer. Cell Rep 14:920–931. https://doi.org/10.1016/j.celrep.2015.12.086.
  • Potente M, Carmeliet P. 2017. The link between angiogenesis and endothelial metabolism. Annu Rev Physiol 79:43–66. https://doi.org/10.1146/annurev-physiol-021115-105134.
  • Xu Y, An X, Guo X, Habtetsion TG, Wang Y, Xu X, Kandala S, Li Q, Li H, Zhang C, Caldwell RB, Fulton DJ, Su Y, Hoda MN, Zhou G, Wu C, Huo Y. 2014. Endothelial PFKFB3 plays a critical role in angiogenesis. Arterioscler Thromb Vasc Biol 34:1231–1239. https://doi.org/10.1161/ATVBAHA.113.303041.
  • Schoors S, Bruning U, Missiaen R, Queiroz KC, Borgers G, Elia I, Zecchin A, Cantelmo AR, Christen S, Goveia J, Heggermont W, Godde L, Vinckier S, Van Veldhoven PP, Eelen G, Schoonjans L, Gerhardt H, Dewerchin M, Baes M, De Bock K, Ghesquiere B, Lunt SY, Fendt SM, Carmeliet P. 2015. Fatty acid carbon is essential for dNTP synthesis in endothelial cells. Nature 520:192–197. https://doi.org/10.1038/nature14362.
  • Kalucka J, Bierhansl L, Conchinha NV, Missiaen R, Elia I, Bruning U, Scheinok S, Treps L, Cantelmo AR, Dubois C, de Zeeuw P, Goveia J, Zecchin A, Taverna F, Morales-Rodriguez F, Brajic A, Conradi LC, Schoors S, Harjes U, Vriens K, Pilz GA, Chen R, Cubbon R, Thienpont B, Cruys B, Wong BW, Ghesquiere B, Dewerchin M, De Bock K, Sagaert X, Jessberger S, Jones EAV, Gallez B, Lambrechts D, Mazzone M, Eelen G, Li X, Fendt SM, Carmeliet P. 2018. Quiescent endothelial cells upregulate fatty acid beta-oxidation for vasculoprotection via redox homeostasis. Cell Metab 28:881–894. https://doi.org/10.1016/j.cmet.2018.07.016.
  • National Research Council. 2011. Guide for the care and use of laboratory animals, 8th ed. National Academies Press, Washington, DC.
  • Yu G, Wang LG, Han Y, He QY. 2012. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics 16:284–287. https://doi.org/10.1089/omi.2011.0118.
  • Eglinger J, Karsjens H, Lammert E. 2017. Quantitative assessment of angiogenesis and pericyte coverage in human cell-derived vascular sprouts. Inflamm Regen 37:016–0033.
  • Carpentier GMM, Courty J, Cascone I. 2012. Angiogenesis analyzer for ImageJ, p 198–201. 4th ImageJ User and Developer Conference Proceedings. Centre de Recherche Henri Tudor, Luxembourg, Belgium.
  • Tual-Chalot S, Allinson KR, Fruttiger M, Arthur HM. 2013. Whole mount immunofluorescent staining of the neonatal mouse retina to investigate angiogenesis in vivo. J Vis Exp 9:50546.
  • Zudaire E, Gambardella L, Kurcz C, Vermeren S. 2011. A computational tool for quantitative analysis of vascular networks. PLoS One 6:e27385. https://doi.org/10.1371/journal.pone.0027385.
  • Langmead B, Salzberg SL. 2012. Fast gapped-read alignment with Bowtie 2. Nat Methods 9:357–359. https://doi.org/10.1038/nmeth.1923.
  • Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum C, Myers RM, Brown M, Li W, Liu XS. 2008. Model-based analysis of ChIP-Seq (MACS). Genome Biol 9:2008–2009.
  • Li QB, James B, Huang H, Bickel PJ. 2011. Measuring reproducibility of high-throughput experiments. Ann Appl Stat 5:1752–1779. https://doi.org/10.1214/11-AOAS466.
  • Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, Cheng JX, Murre C, Singh H, Glass CK. 2010. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38:576–589. https://doi.org/10.1016/j.molcel.2010.05.004.
  • Wang J, Zhuang J, Iyer S, Lin X, Whitfield TW, Greven MC, Pierce BG, Dong X, Kundaje A, Cheng Y, Rando OJ, Birney E, Myers RM, Noble WS, Snyder M, Weng Z. 2012. Sequence features and chromatin structure around the genomic regions bound by 119 human transcription factors. Genome Res 22:1798–1812. https://doi.org/10.1101/gr.139105.112.
  • Ramirez F, Ryan DP, Gruning B, Bhardwaj V, Kilpert F, Richter AS, Heyne S, Dundar F, Manke T. 2016. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res 44:13.
  • Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G. 2000. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25:25–29. https://doi.org/10.1038/75556.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.