Skip to main content

Advertisement

Log in

Epigenetic Regulation of Vascular Endothelial Biology/Pathobiology and Response to Fluid Shear Stress

  • Published:
Cellular and Molecular Bioengineering Aims and scope Submit manuscript

Abstract

Vascular endothelial cells (ECs) are constantly exposed to hemodynamic forces, including blood flow-induced shear stress, which modulates EC gene expression and function and hence vascular biology/pathobiology in health and disease. Epigenetics refers to chromatin-based mechanisms, including DNA methylation, histone modifications, and RNA-based machinery, which regulate gene expression without changes in the underlying DNA sequences. The role of epigenetic mechanisms in regulating EC gene expression and function under static condition and in response to shear stress has recently emerged. This review provides an introduction to epigenetic concepts for vascular bioengineers and biologists. Using endothelial nitric oxide synthase, angiogenesis, and atherogenesis as examples, this review presents a conceptual framework for understanding how epigenetic factors, including histone deacetylases and microRNAs, are involved in the control of EC gene expression and function and hence vascular disease development, and summarizes the current knowledge on the role of epigenetic pathways in regulating EC responses to shear stress. Such information can contribute to our understanding of how mechanical environment of ECs impacts their genome to modify disease susceptibility and help to generate new approaches for therapeutic interventions.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1
Figure 2
Figure 3

Similar content being viewed by others

Abbreviations

3′-UTR:

3′-untranslated region.

AP-1:

activator protein-1

apoE−/− :

apolipoprotein E-deficient

cFLIP:

cellular caspase-8 (FLICE)-like inhibitory protein

DNMT:

DNA methyltransferase

EC:

endothelial cell

eNOS:

endothelial nitric oxide synthase

ER:

estrogen receptor

ERK:

extracellular-regulated kinase

FGF:

fibroblast growth factor

FoxO1:

forkhead box protein O1

HAT:

histone acetyltransferase

HDAC:

histone deacetylase

HMT:

histone N-methyltransferase

HUVEC:

human umbilical vein endothelial cell

ICAM-1:

intercellular adhesion molecule-1

IL:

interleukin

KLF-2:

krüppel-like factor 2

LDL:

low density lipoprotein

LSS:

laminar shear stress

MAPK:

mitogen-activated protein kinase

MBD:

methyl-CpG-binding domain

MCP-1:

monocyte chemotactic protein-1

MeCP2:

methyl CpG binding protein 2

MEF-2:

myocyte enhancer factor-2

miR:

microRNA

ncRNA:

noncoding RNA

NF-κB:

nuclear factor-κB

NO:

nitric oxide

OSS:

oscillatory shear stress

oxLDL:

oxide low density lipoprotein

PI3K:

phosphoinositide-3-kinase

PPAR:

peroxisome proliferators-activated receptor

pre-miR:

precursor microRNA

pri-miR:

primary microRNA

PSS:

pulsatile shear stress

SIRT:

sirtuin, information regulator

Sir2:

information regulator 2

SMC:

smooth muscle cell

STAT5A:

signal transducer and activator of transcription 5A

TNF-α:

tumor necrosis factor-α

TSA:

trichostatin A

VCAM-1:

vascular adhesion molecule-1

VEGF:

vascular endothelial growth factor

References

  1. Aitsebaomo, J., A. L. Portbury, J. C. Schisler, and C. Patterson. Brothers and sisters: molecular insights into arterial–venous heterogeneity. Circ. Res. 103(9):929–939, 2008.

    Google Scholar 

  2. Aurora, A. B., D. Biyashev, Y. Mirochnik, T. A. Zaichuk, C. Sanchez-Martinez, M. A. Renault, D. Losordo, and O. V. Volpert. NF-kappaB balances vascular regression and angiogenesis via chromatin remodeling and NFAT displacement. Blood 116(3):475–484, 2010.

    Google Scholar 

  3. Azumi, H., N. Inoue, S. Takeshita, Y. Rikitake, S. Kawashima, Y. Hayashi, H. Itoh, and M. Yokoyama. Expression of NADH/NADPH oxidase p22phox in human coronary arteries. Circulation 100(14):1494–1498, 1999.

    Google Scholar 

  4. Baldwin, K. M., and F. Haddad. Research in the exercise sciences: where we are and where do we go from here. Part II. Exerc. Sport Sci. Rev. 38(2):42–50, 2010.

    Google Scholar 

  5. Bannister, A. J., and T. Kouzarides. Histone methylation: recognizing the methyl mark. Methods Enzymol. 376:269–288, 2004.

    Google Scholar 

  6. Bannister, A. J., and T. Kouzarides. Reversing histone methylation. Nature 436(7054):1103–1106, 2005.

    Google Scholar 

  7. Barski, A., S. Cuddapah, K. Cui, T. Y. Roh, D. E. Schones, Z. Wang, G. Wei, I. Chepelev, and K. Zhao. High-resolution profiling of histone methylations in the human genome. Cell 129(4):823–837, 2007.

    Google Scholar 

  8. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136(2):215–233, 2009.

    Google Scholar 

  9. Bavan, L., K. Midwood, and J. Nanchahal. MicroRNA epigenetics: a new avenue for wound healing research. BioDrugs 25(1):27–41, 2011.

    Google Scholar 

  10. Baylin, S. B. DNA methylation and gene silencing in cancer. Nat. Clin. Pract. Oncol. 2(Suppl 1):S4–S11, 2005.

    Google Scholar 

  11. Berk, B. C. Atheroprotective signaling mechanisms activated by steady laminar flow in endothelial cells. Circulation 117(8):1082–1089, 2008.

    Google Scholar 

  12. Bernstein, B. E., A. Meissner, and E. S. Lander. The mammalian epigenome. Cell 128(4):669–681, 2007.

    Google Scholar 

  13. Bonauer, A., G. Carmona, M. Iwasaki, M. Mione, M. Koyanagi, A. Fischer, J. Burchfield, H. Fox, C. Doebele, K. Ohtani, E. Chavakis, M. Potente, M. Tjwa, C. Urbich, A. M. Zeiher, and S. Dimmeler. MicroRNA-92a controls angiogenesis and functional recovery of ischemic tissues in mice. Science 324(5935):1710–1713, 2009.

    Google Scholar 

  14. Buga, G. M., M. E. Gold, J. M. Fukuto, and L. J. Ignarro. Shear stress-induced release of nitric oxide from endothelial cells grown on beads. Hypertension 17(2):187–193, 1991.

    Google Scholar 

  15. Burdge, G. C., M. A. Hanson, J. L. Slater-Jefferies, and K. A. Lillycrop. Epigenetic regulation of transcription: a mechanism for inducing variations in phenotype (fetal programming) by differences in nutrition during early life? Br. J. Nutr. 97(6):1036–1046, 2007.

    Google Scholar 

  16. Buysschaert, I., T. Schmidt, C. Roncal, P. Carmeliet, and D. Lambrechts. Genetics, epigenetics and pharmaco-(epi)genomics in angiogenesis. J. Cell. Mol. Med. 12(6B):2533–2551, 2008.

    Google Scholar 

  17. Carthew, R. W., and E. J. Sontheimer. Origins and Mechanisms of miRNAs and siRNAs. Cell 136(4):642–655, 2009.

    Google Scholar 

  18. Chan, L. S., P. Y. Yue, N. K. Mak, and R. N. Wong. Role of microRNA-214 in ginsenoside-Rg1-induced angiogenesis. Eur. J. Pharm. Sci. 38(4):370–377, 2009.

    Google Scholar 

  19. Chan, Y., J. E. Fish, C. D’Abreo, S. Lin, G. B. Robb, A. M. Teichert, F. Karantzoulis-Fegaras, A. Keightley, B. M. Steer, and P. A. Marsden. The cell-specific expression of endothelial nitric-oxide synthase: a role for DNA methylation. J. Biol. Chem. 279(33):35087–35100, 2004.

    Google Scholar 

  20. Chen, Y., and D. H. Gorski. Regulation of angiogenesis through a microRNA (miR-130a) that down-regulates antiangiogenic homeobox genes GAX and HOXA5. Blood 111(3):1217–1226, 2008.

    Google Scholar 

  21. Chen, Z., I. C. Peng, X. Cui, Y. S. Li, S. Chien, and J. Y. Shyy. Shear stress, SIRT1, and vascular homeostasis. Proc. Natl Acad. Sci. USA 107(22):10268–10273, 2010.

    Google Scholar 

  22. Cheng, C., D. Tempel, R. van Haperen, A. van der Baan, F. Grosveld, M. J. Daemen, R. Krams, and R. de Crom. Atherosclerotic lesion size and vulnerability are determined by patterns of fluid shear stress. Circulation 113(23):2744–2753, 2006.

    Google Scholar 

  23. Chien, S. Molecular and mechanical bases of focal lipid accumulation in arterial wall. Prog. Biophys. Mol. Biol. 83(2):131–151, 2003.

    Google Scholar 

  24. Chien, S. Mechanotransduction and endothelial cell homeostasis: the wisdom of the cell. Am. J. Physiol. Heart Circ. Physiol. 292(3):H1209–H1224, 2007.

    Google Scholar 

  25. Chien, S., S. Usami, J. Dellenback, and V. Magazinovic. Blood rheology after hemorrhage and endotoxin. Adv. Exp. Med. Biol. 33:75–93, 1972.

    Google Scholar 

  26. Chiu, J. J., and S. Chien. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol. Rev. 91(1):327–387, 2011.

    Google Scholar 

  27. Chiu, J. J., S. Usami, and S. Chien. Vascular endothelial responses to altered shear stress: pathologic implications for atherosclerosis. Ann. Med. 41(1):19–28, 2009.

    Google Scholar 

  28. Cooke, J. P. Flow, NO, and atherogenesis. Proc. Natl Acad. Sci. USA 100(3):768–770, 2003.

    Google Scholar 

  29. Davies, P. F. Flow-mediated endothelial mechanotransduction. Physiol. Rev. 75(3):519–560, 1995.

    Google Scholar 

  30. de Nigris, F., L. O. Lerman, S. W. Ignarro, G. Sica, A. Lerman, W. Palinski, L. J. Ignarro, and C. Napoli. Beneficial effects of antioxidants and l-arginine on oxidation-sensitive gene expression and endothelial NO synthase activity at sites of disturbed shear stress. Proc. Natl Acad. Sci. USA 100(3):1420–1425, 2003.

    Google Scholar 

  31. Dentelli, P., A. Rosso, F. Orso, C. Olgasi, D. Taverna, and M. F. Brizzi. microRNA-222 controls neovascularization by regulating signal transducer and activator of transcription 5A expression. Arterioscler. Thromb. Vasc. Biol. 30(8):1562–1568, 2010.

    Google Scholar 

  32. Deroanne, C. F., K. Bonjean, S. Servotte, L. Devy, A. Colige, N. Clausse, S. Blacher, E. Verdin, J. M. Foidart, B. V. Nusgens, and V. Castronovo. Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene 21(3):427–436, 2002.

    Google Scholar 

  33. Dje N’Guessan, P., F. Riediger, K. Vardarova, S. Scharf, J. Eitel, B. Opitz, H. Slevogt, W. Weichert, A. C. Hocke, B. Schmeck, N. Suttorp, and S. Hippenstiel. Statins control oxidized LDL-mediated histone modifications and gene expression in cultured human endothelial cells. Arterioscler. Thromb. Vasc. Biol. 29(3):380–386, 2009.

    Google Scholar 

  34. Doebele, C., A. Bonauer, A. Fischer, A. Scholz, Y. Reiss, C. Urbich, W. K. Hofmann, A. M. Zeiher, and S. Dimmeler. Members of the microRNA-17–92 cluster exhibit a cell-intrinsic antiangiogenic function in endothelial cells. Blood 115(23):4944–4950, 2010.

    Google Scholar 

  35. Drexler, H., and B. Hornig. Endothelial dysfunction in human disease. J. Mol. Cell. Cardiol. 31(1):51–60, 1999.

    Google Scholar 

  36. Duncker, D. J., and R. J. Bache. Regulation of coronary blood flow during exercise. Physiol. Rev. 88(3):1009–1086, 2008.

    Google Scholar 

  37. Fang, Y., C. Shi, E. Manduchi, M. Civelek, and P. F. Davies. MicroRNA-10a regulation of proinflammatory phenotype in athero-susceptible endothelium in vivo and in vitro. Proc. Natl Acad. Sci. USA 107(30):13450–13455, 2010.

    Google Scholar 

  38. Feletou, M., and P. M. Vanhoutte. Endothelial dysfunction: a multifaceted disorder (The Wiggers Award Lecture). Am. J. Physiol. Heart Circ. Physiol. 291(3):H985–H1002, 2006.

    Google Scholar 

  39. Feletou, M., and P. M. Vanhoutte. Endothelium-derived hyperpolarizing factor: Where are we now? Arterioscler. Thromb. Vasc. Biol. 26(6):1215–1225, 2006.

    Google Scholar 

  40. Fernandez, A. Z., A. L. Siebel, and A. El-Osta. Atherogenic factors and their epigenetic relationships. Int. J. Vasc. Med. 2010:437809, 2010.

    Google Scholar 

  41. Fish, J. E., C. C. Matouk, A. Rachlis, S. Lin, S. C. Tai, C. D’Abreo, and P. A. Marsden. The expression of endothelial nitric-oxide synthase is controlled by a cell-specific histone code. J. Biol. Chem. 280(26):24824–24838, 2005.

    Google Scholar 

  42. Fish, J. E., M. M. Santoro, S. U. Morton, S. Yu, R. F. Yeh, J. D. Wythe, K. N. Ivey, B. G. Bruneau, D. Y. Stainier, and D. Srivastava. miR-126 regulates angiogenic signaling and vascular integrity. Dev. Cell. 15(2):272–284, 2008.

    Google Scholar 

  43. Fuks, F., P. J. Hurd, D. Wolf, X. Nan, A. P. Bird, and T. Kouzarides. The methyl-CpG-binding protein MeCP2 links DNA methylation to histone methylation. J. Biol. Chem. 278(6):4035–4040, 2003.

    Google Scholar 

  44. Gambillara, V., C. Chambaz, G. Montorzi, S. Roy, N. Stergiopulos, and P. Silacci. Plaque-prone hemodynamics impair endothelial function in pig carotid arteries. Am. J. Physiol. Heart Circ. Physiol. 290(6):H2320–H2328, 2006.

    Google Scholar 

  45. Gan, Y., Y. H. Shen, J. Wang, X. Wang, B. Utama, and X. L. Wang. Role of histone deacetylation in cell-specific expression of endothelial nitric-oxide synthase. J. Biol. Chem. 280(16):16467–16475, 2005.

    Google Scholar 

  46. Garin, G., and B. C. Berk. Flow-mediated signaling modulates endothelial cell phenotype. Endothelium 13(6):375–384, 2006.

    Google Scholar 

  47. Gibbons, G. H., and V. J. Dzau. The emerging concept of vascular remodeling. N. Engl. J. Med. 330(20):1431–1438, 1994.

    Google Scholar 

  48. Gimbrone, Jr., M. A. Vascular endothelium, hemodynamic forces, and atherogenesis. Am. J. Pathol. 155(1):1–5, 1999.

    Google Scholar 

  49. Goligorsky, M. S., and T. Rabelink. Meeting report: ISN forefronts in nephrology on endothelial biology and renal disease: from bench to prevention. Kidney Int. 70(2):258–264, 2006.

    Google Scholar 

  50. Grundmann, S., F. P. Hans, S. Kinniry, J. Heinke, T. Helbing, F. Bluhm, J. P. Sluijter, I. Hoefer, G. Pasterkamp, C. Bode, and M. Moser. MicroRNA-100 regulates neovascularization by suppression of mammalian target of rapamycin in endothelial and vascular smooth muscle cells. Circulation 123(9):999–1009, 2011.

    Google Scholar 

  51. Harris, R. A., T. Wang, C. Coarfa, R. P. Nagarajan, C. Hong, S. L. Downey, B. E. Johnson, S. D. Fouse, A. Delaney, Y. Zhao, A. Olshen, T. Ballinger, X. Zhou, K. J. Forsberg, J. Gu, L. Echipare, H. O’Geen, R. Lister, M. Pelizzola, Y. Xi, C. B. Epstein, B. E. Bernstein, R. D. Hawkins, B. Ren, W. Y. Chung, H. Gu, C. Bock, A. Gnirke, M. Q. Zhang, D. Haussler, J. R. Ecker, W. Li, P. J. Farnham, R. A. Waterland, A. Meissner, M. A. Marra, M. Hirst, A. Milosavljevic, and J. F. Costello. Comparison of sequencing-based methods to profile DNA methylation and identification of monoallelic epigenetic modifications. Nat. Biotechnol. 28(10):1097–1105, 2010.

    Google Scholar 

  52. Harris, T. A., M. Yamakuchi, M. Ferlito, J. T. Mendell, and C. J. Lowenstein. MicroRNA-126 regulates endothelial expression of vascular cell adhesion molecule 1. Proc. Natl Acad. Sci. USA 105(5):1516–1521, 2008.

    Google Scholar 

  53. Harrison, D. G. Cellular and molecular mechanisms of endothelial cell dysfunction. J. Clin. Invest. 100(9):2153–2157, 1997.

    Google Scholar 

  54. Harrison, D. G. The shear stress of keeping arteries clear. Nat. Med. 11(4):375–376, 2005.

    Google Scholar 

  55. Harrison, D. G., J. Widder, I. Grumbach, W. Chen, M. Weber, and C. Searles. Endothelial mechanotransduction, nitric oxide and vascular inflammation. J. Intern. Med. 259(4):351–363, 2006.

    Google Scholar 

  56. Hebbel, R. P., G. M. Vercellotti, B. S. Pace, A. N. Solovey, R. Kollander, C. F. Abanonu, J. Nguyen, J. V. Vineyard, J. D. Belcher, F. Abdulla, S. Osifuye, J. W. Eaton, R. J. Kelm, Jr., and A. Slungaard. The HDAC inhibitors trichostatin A and suberoylanilide hydroxamic acid exhibit multiple modalities of benefit for the vascular pathobiology of sickle transgenic mice. Blood 115(12):2483–2490, 2010.

    Google Scholar 

  57. Hendrich, B., and S. Tweedie. The methyl-CpG binding domain and the evolving role of DNA methylation in animals. Trends Genet. 19(5):269–277, 2003.

    Google Scholar 

  58. Hiltunen, M. O., M. P. Turunen, T. P. Hakkinen, J. Rutanen, M. Hedman, K. Makinen, A. M. Turunen, K. Aalto-Setala, and S. Yla-Herttuala. DNA hypomethylation and methyltransferase expression in atherosclerotic lesions. Vasc. Med. 7(1):5–11, 2002.

    Google Scholar 

  59. Illi, B., C. Dello Russo, C. Colussi, J. Rosati, M. Pallaoro, F. Spallotta, D. Rotili, S. Valente, G. Ragone, F. Martelli, P. Biglioli, C. Steinkuhler, P. Gallinari, A. Mai, M. C. Capogrossi, and C. Gaetano. Nitric oxide modulates chromatin folding in human endothelial cells via protein phosphatase 2A activation and class II histone deacetylases nuclear shuttling. Circ. Res. 102(1):51–58, 2008.

    Google Scholar 

  60. Illi, B., S. Nanni, A. Scopece, A. Farsetti, P. Biglioli, M. C. Capogrossi, and C. Gaetano. Shear stress-mediated chromatin remodeling provides molecular basis for flow-dependent regulation of gene expression. Circ. Res. 93(2):155–161, 2003.

    Google Scholar 

  61. Inoue, K., M. Kobayashi, K. Yano, M. Miura, A. Izumi, C. Mataki, T. Doi, T. Hamakubo, P. C. Reid, D. A. Hume, M. Yoshida, W. C. Aird, T. Kodama, and T. Minami. Histone deacetylase inhibitor reduces monocyte adhesion to endothelium through the suppression of vascular cell adhesion molecule-1 expression. Arterioscler. Thromb. Vasc. Biol. 26(12):2652–2659, 2006.

    Google Scholar 

  62. Ito, T., S. Yagi, and M. Yamakuchi. MicroRNA-34a regulation of endothelial senescence. Biochem. Biophys. Res. Commun. 398(4):735–740, 2010.

    Google Scholar 

  63. Ji, R., Y. Cheng, J. Yue, J. Yang, X. Liu, H. Chen, D. B. Dean, and C. Zhang. MicroRNA expression signature and antisense-mediated depletion reveal an essential role of MicroRNA in vascular neointimal lesion formation. Circ. Res. 100(11):1579–1588, 2007.

    Google Scholar 

  64. Kamiya, A., and T. Togawa. Adaptive regulation of wall shear stress to flow change in the canine carotid artery. Am. J. Physiol. 239(1):H14–H21, 1980.

    Google Scholar 

  65. Kim, J., J. Y. Kim, K. S. Song, Y. H. Lee, J. S. Seo, J. Jelinek, P. J. Goldschmidt-Clermont, and J. P. Issa. Epigenetic changes in estrogen receptor beta gene in atherosclerotic cardiovascular tissues and in vitro vascular senescence. Biochim. Biophys. Acta 1772(1):72–80, 2007.

    Google Scholar 

  66. Kim, M. S., H. J. Kwon, Y. M. Lee, J. H. Baek, J. E. Jang, S. W. Lee, E. J. Moon, H. S. Kim, S. K. Lee, H. Y. Chung, C. W. Kim, and K. W. Kim. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat. Med. 7(4):437–443, 2001.

    Google Scholar 

  67. Kouzarides, T. Histone methylation in transcriptional control. Curr. Opin. Genet. Dev. 12(2):198–209, 2002.

    Google Scholar 

  68. Kouzarides, T. Chromatin modifications and their function. Cell 128(4):693–705, 2007.

    Google Scholar 

  69. Langille, B. L., and F. O’Donnell. Reductions in arterial diameter produced by chronic decreases in blood flow are endothelium-dependent. Science 231(4736):405–407, 1986.

    Google Scholar 

  70. Laukkanen, M. O., S. Mannermaa, M. O. Hiltunen, S. Aittomaki, K. Airenne, J. Janne, and S. Yla-Herttuala. Local hypomethylation in atherosclerosis found in rabbit ec-sod gene. Arterioscler. Thromb. Vasc. Biol. 19(9):2171–2178, 1999.

    Google Scholar 

  71. Li, E., T. H. Bestor, and R. Jaenisch. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69(6):915–926, 1992.

    Google Scholar 

  72. Li, Y., M. A. Reddy, F. Miao, N. Shanmugam, J. K. Yee, D. Hawkins, B. Ren, and R. Natarajan. Role of the histone H3 lysine 4 methyltransferase, SET7/9, in the regulation of NF-kappaB-dependent inflammatory genes. Relevance to diabetes and inflammation. J. Biol. Chem. 283(39):26771–26781, 2008.

    Google Scholar 

  73. Li, Y. S., J. H. Haga, and S. Chien. Molecular basis of the effects of shear stress on vascular endothelial cells. J. Biomech. 38(10):1949–1971, 2005.

    Google Scholar 

  74. Lin, X., and W. G. Nelson. Methyl-CpG-binding domain protein-2 mediates transcriptional repression associated with hypermethylated GSTP1 CpG islands in MCF-7 breast cancer cells. Cancer Res. 63(2):498–504, 2003.

    Google Scholar 

  75. Lund, G., L. Andersson, M. Lauria, M. Lindholm, M. F. Fraga, A. Villar-Garea, E. Ballestar, M. Esteller, and S. Zaina. DNA methylation polymorphisms precede any histological sign of atherosclerosis in mice lacking apolipoprotein E. J. Biol. Chem. 279(28):29147–29154, 2004.

    Google Scholar 

  76. Lusser, A., and J. T. Kadonaga. Chromatin remodeling by ATP-dependent molecular machines. Bioessays 25(12):1192–1200, 2003.

    Google Scholar 

  77. Malek, A. M., S. L. Alper, and S. Izumo. Hemodynamic shear stress and its role in atherosclerosis. JAMA 282(21):2035–2042, 1999.

    Google Scholar 

  78. Margariti, A., A. Zampetaki, Q. Xiao, B. Zhou, E. Karamariti, D. Martin, X. Yin, M. Mayr, H. Li, Z. Zhang, E. De Falco, Y. Hu, G. Cockerill, Q. Xu, and L. Zeng. Histone deacetylase 7 controls endothelial cell growth through modulation of beta-catenin. Circ. Res. 106(7):1202–1211, 2010.

    Google Scholar 

  79. Marmorstein, R. Protein modules that manipulate histone tails for chromatin regulation. Nat. Rev. Mol. Cell Biol. 2(6):422–432, 2001.

    Google Scholar 

  80. Matouk, C. C., and P. A. Marsden. Epigenetic regulation of vascular endothelial gene expression. Circ. Res. 102(8):873–887, 2008.

    Google Scholar 

  81. Mattagajasingh, I., C. S. Kim, A. Naqvi, T. Yamamori, T. A. Hoffman, S. B. Jung, J. DeRicco, K. Kasuno, and K. Irani. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc. Natl Acad. Sci. USA 104(37):14855–14860, 2007.

    Google Scholar 

  82. Mattick, J. S. The genetic signatures of noncoding RNAs. PLoS Genet. 5(4):e1000459, 2009.

    Google Scholar 

  83. McLenachan, J. M., J. Vita, D. R. Fish, C. B. Treasure, D. A. Cox, P. Ganz, and A. P. Selwyn. Early evidence of endothelial vasodilator dysfunction at coronary branch points. Circulation 82(4):1169–1173, 1990.

    Google Scholar 

  84. Menghini, R., V. Casagrande, M. Cardellini, E. Martelli, A. Terrinoni, F. Amati, M. Vasa-Nicotera, A. Ippoliti, G. Novelli, G. Melino, R. Lauro, and M. Federici. MicroRNA 217 modulates endothelial cell senescence via silent information regulator 1. Circulation 120(15):1524–1532, 2009.

    Google Scholar 

  85. Minor, R. L., Jr., P. R. Myers, R. Guerra, Jr., J. N. Bates, and D. G. Harrison. Diet-induced atherosclerosis increases the release of nitrogen oxides from rabbit aorta. J. Clin. Invest. 86(6):2109–2116, 1990.

    Google Scholar 

  86. Miranda, T. B., and P. A. Jones. DNA methylation: the nuts and bolts of repression. J. Cell. Physiol. 213(2):384–390, 2007.

    Google Scholar 

  87. Mottet, D., A. Bellahcene, S. Pirotte, D. Waltregny, C. Deroanne, V. Lamour, R. Lidereau, and V. Castronovo. Histone deacetylase 7 silencing alters endothelial cell migration, a key step in angiogenesis. Circ. Res. 101(12):1237–1246, 2007.

    Google Scholar 

  88. Mueller, M. A., F. Beutner, D. Teupser, U. Ceglarek, and J. Thiery. Prevention of atherosclerosis by the mTOR inhibitor everolimus in LDLR−/− mice despite severe hypercholesterolemia. Atherosclerosis 198(1):39–48, 2008.

    Google Scholar 

  89. Nakashima, Y., A. S. Plump, E. W. Raines, J. L. Breslow, and R. Ross. ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler. Thromb. 14(1):133–140, 1994.

    Google Scholar 

  90. Nerem, R. M., and D. Seliktar. Vascular tissue engineering. Annu. Rev. Biomed. Eng. 3:225–243, 2001.

    Google Scholar 

  91. Ni, C. W., H. Qiu, and H. Jo. MicroRNA-663 upregulated by oscillatory shear stress plays a role in inflammatory response of endothelial cells. Am. J. Physiol. Heart Circ. Physiol. 300(5):H1762–H1769, 2011.

    Google Scholar 

  92. Noris, M., M. Morigi, R. Donadelli, S. Aiello, M. Foppolo, M. Todeschini, S. Orisio, G. Remuzzi, and A. Remuzzi. Nitric oxide synthesis by cultured endothelial cells is modulated by flow conditions. Circ. Res. 76(4):536–543, 1995.

    Google Scholar 

  93. Okano, M., D. W. Bell, D. A. Haber, and E. Li. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99(3):247–257, 1999.

    Google Scholar 

  94. Olesen, S. P., D. E. Clapham, and P. F. Davies. Haemodynamic shear stress activates a K+ current in vascular endothelial cells. Nature 331(6152):168–170, 1988.

    Google Scholar 

  95. Ollikainen, M., K. R. Smith, E. J. Joo, H. K. Ng, R. Andronikos, B. Novakovic, N. K. Abdul Aziz, J. B. Carlin, R. Morley, R. Saffery, and J. M. Craig. DNA methylation analysis of multiple tissues from newborn twins reveals both genetic and intrauterine components to variation in the human neonatal epigenome. Hum. Mol. Genet. 19(21):4176–4188, 2010.

    Google Scholar 

  96. Ordovas, J. M., and C. E. Smith. Epigenetics and cardiovascular disease. Nat. Rev. Cardiol. 7(9):510–519, 2010.

    Google Scholar 

  97. Pohl, U., C. De Wit, and T. Gloe. Large arterioles in the control of blood flow: role of endothelium-dependent dilation. Acta Physiol. Scand. 168(4):505–510, 2000.

    Google Scholar 

  98. Potente, M., L. Ghaeni, D. Baldessari, R. Mostoslavsky, L. Rossig, F. Dequiedt, J. Haendeler, M. Mione, E. Dejana, F. W. Alt, A. M. Zeiher, and S. Dimmeler. SIRT1 controls endothelial angiogenic functions during vascular growth. Genes Dev. 21(20):2644–2658, 2007.

    Google Scholar 

  99. Qin, X., X. Wang, Y. Wang, Z. Tang, Q. Cui, J. Xi, Y. S. Li, S. Chien, and N. Wang. MicroRNA-19a mediates the suppressive effect of laminar flow on cyclin D1 expression in human umbilical vein endothelial cells. Proc. Natl Acad. Sci. USA 107(7):3240–3244, 2010.

    Google Scholar 

  100. Risau, W. Mechanisms of angiogenesis. Nature 386(6626):671–674, 1997.

    Google Scholar 

  101. Rossig, L., H. Li, B. Fisslthaler, C. Urbich, I. Fleming, U. Forstermann, A. M. Zeiher, and S. Dimmeler. Inhibitors of histone deacetylation downregulate the expression of endothelial nitric oxide synthase and compromise endothelial cell function in vasorelaxation and angiogenesis. Circ. Res. 91(9):837–844, 2002.

    Google Scholar 

  102. Rossig, L., C. Urbich, T. Bruhl, E. Dernbach, C. Heeschen, E. Chavakis, K. Sasaki, D. Aicher, F. Diehl, F. Seeger, M. Potente, A. Aicher, L. Zanetta, E. Dejana, A. M. Zeiher, and S. Dimmeler. Histone deacetylase activity is essential for the expression of HoxA9 and for endothelial commitment of progenitor cells. J. Exp. Med. 201(11):1825–1835, 2005.

    Google Scholar 

  103. Schones, D. E., K. Cui, S. Cuddapah, T. Y. Roh, A. Barski, Z. Wang, G. Wei, and K. Zhao. Dynamic regulation of nucleosome positioning in the human genome. Cell 132(5):887–898, 2008.

    Google Scholar 

  104. Searles, C. D. Transcriptional and posttranscriptional regulation of endothelial nitric oxide synthase expression. Am. J. Physiol. Cell Physiol. 291(5):C803–C816, 2006.

    Google Scholar 

  105. Shahbazian, M. D., and M. Grunstein. Functions of site-specific histone acetylation and deacetylation. Annu. Rev. Biochem. 76:75–100, 2007.

    Google Scholar 

  106. Sincic, N., and Z. Herceg. DNA methylation and cancer: ghosts and angels above the genes. Curr. Opin. Oncol. 23(1):69–76, 2011.

    Google Scholar 

  107. Siomi, H., and M. C. Siomi. Posttranscriptional regulation of microRNA biogenesis in animals. Mol. Cell. 38(3):323–332, 2010.

    Google Scholar 

  108. Stein, S., and C. M. Matter. Protective roles of SIRT1 in atherosclerosis. Cell Cycle 10(4):640–647, 2011.

    Google Scholar 

  109. Suarez, Y., C. Fernandez-Hernando, J. S. Pober, and W. C. Sessa. Dicer dependent microRNAs regulate gene expression and functions in human endothelial cells. Circ. Res. 100(8):1164–1173, 2007.

    Google Scholar 

  110. Suarez, Y., C. Fernandez-Hernando, J. Yu, S. A. Gerber, K. D. Harrison, J. S. Pober, M. L. Iruela-Arispe, M. Merkenschlager, and W. C. Sessa. Dicer-dependent endothelial microRNAs are necessary for postnatal angiogenesis. Proc. Natl Acad. Sci. USA 105(37):14082–14087, 2008.

    Google Scholar 

  111. Suarez, Y., and W. C. Sessa. MicroRNAs as novel regulators of angiogenesis. Circ. Res. 104(4):442–454, 2009.

    Google Scholar 

  112. Sun, W., Y. S. Julie Li, H. D. Huang, J. Y. Shyy, and S. Chien. microRNA: a master regulator of cellular processes for bioengineering systems. Annu. Rev. Biomed. Eng. 12:1–27, 2010.

    Google Scholar 

  113. Taganov, K. D., M. P. Boldin, K. J. Chang, and D. Baltimore. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl Acad. Sci. USA 103(33):12481–12486, 2006.

    Google Scholar 

  114. Tai, S. C., G. B. Robb, and P. A. Marsden. Endothelial nitric oxide synthase: a new paradigm for gene regulation in the injured blood vessel. Arterioscler. Thromb. Vasc. Biol. 24(3):405–412, 2004.

    Google Scholar 

  115. Topper, J. N., and M. A. Gimbrone, Jr. Blood flow and vascular gene expression: fluid shear stress as a modulator of endothelial phenotype. Mol. Med. Today 5(1):40–46, 1999.

    Google Scholar 

  116. Traub, O., and B. C. Berk. Laminar shear stress: mechanisms by which endothelial cells transduce an atheroprotective force. Arterioscler. Thromb. Vasc. Biol. 18(5):677–685, 1998.

    Google Scholar 

  117. Turunen, M. P., E. Aavik, and S. Yla-Herttuala. Epigenetics and atherosclerosis. Biochim. Biophys. Acta 1790(9):886–891, 2009.

    Google Scholar 

  118. Uematsu, M., Y. Ohara, J. P. Navas, K. Nishida, T. J. Murphy, R. W. Alexander, R. M. Nerem, and D. G. Harrison. Regulation of endothelial cell nitric oxide synthase mRNA expression by shear stress. Am. J. Physiol. 269(6 Pt 1):C1371–C1378, 1995.

    Google Scholar 

  119. Urbich, C., L. Rossig, D. Kaluza, M. Potente, J. N. Boeckel, A. Knau, F. Diehl, J. G. Geng, W. K. Hofmann, A. M. Zeiher, and S. Dimmeler. HDAC5 is a repressor of angiogenesis and determines the angiogenic gene expression pattern of endothelial cells. Blood 113(22):5669–5679, 2009.

    Google Scholar 

  120. Utley, R. T., K. Ikeda, P. A. Grant, J. Cote, D. J. Steger, A. Eberharter, S. John, and J. L. Workman. Transcriptional activators direct histone acetyltransferase complexes to nucleosomes. Nature 394(6692):498–502, 1998.

    Google Scholar 

  121. Vaissiere, T., C. Sawan, and Z. Herceg. Epigenetic interplay between histone modifications and DNA methylation in gene silencing. Mutat. Res. 659(1–2):40–48, 2008.

    Google Scholar 

  122. Wang, G. G., C. D. Allis, and P. Chi. Chromatin remodeling and cancer. Part I. Covalent histone modifications. Trends Mol. Med. 13(9):363–372, 2007.

    Google Scholar 

  123. Wang, J., S. A. Mahmud, P. B. Bitterman, Y. Huo, and A. Slungaard. Histone deacetylase inhibitors suppress TF-kappaB-dependent agonist-driven tissue factor expression in endothelial cells and monocytes. J. Biol. Chem. 282(39):28408–28418, 2007.

    Google Scholar 

  124. Wang, K. C., L. X. Garmire, A. Young, P. Nguyen, A. Trinh, S. Subramaniam, N. Wang, J. Y. Shyy, Y. S. Li, and S. Chien. Role of microRNA-23b in flow-regulation of Rb phosphorylation and endothelial cell growth. Proc. Natl Acad. Sci. USA 107(7):3234–3239, 2010.

    Google Scholar 

  125. Wang, S., A. B. Aurora, B. A. Johnson, X. Qi, J. McAnally, J. A. Hill, J. A. Richardson, R. Bassel-Duby, and E. N. Olson. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev. Cell. 15(2):261–271, 2008.

    Google Scholar 

  126. Wang, S., and E. N. Olson. AngiomiRs—key regulators of angiogenesis. Curr. Opin. Genet. Dev. 19(3):205–211, 2009.

    Google Scholar 

  127. Wang, W., C. H. Ha, B. S. Jhun, C. Wong, M. K. Jain, and Z. G. Jin. Fluid shear stress stimulates phosphorylation-dependent nuclear export of HDAC5 and mediates expression of KLF2 and eNOS. Blood 115(14):2971–2979, 2010.

    Google Scholar 

  128. Ward, M. R., G. Pasterkamp, A. C. Yeung, and C. Borst. Arterial remodeling. Mechanisms and clinical implications. Circulation 102(10):1186–1191, 2000.

    Google Scholar 

  129. Warnholtz, A., G. Nickenig, E. Schulz, R. Macharzina, J. H. Brasen, M. Skatchkov, T. Heitzer, J. P. Stasch, K. K. Griendling, D. G. Harrison, M. Bohm, T. Meinertz, and T. Munzel. Increased NADH-oxidase-mediated superoxide production in the early stages of atherosclerosis: evidence for involvement of the renin-angiotensin system. Circulation 99(15):2027–2033, 1999.

    Google Scholar 

  130. Weaver, I. C., F. A. Champagne, S. E. Brown, S. Dymov, S. Sharma, M. J. Meaney, and M. Szyf. Reversal of maternal programming of stress responses in adult offspring through methyl supplementation: altering epigenetic marking later in life. J. Neurosci. 25(47):11045–11054, 2005.

    Google Scholar 

  131. Weber, M., M. B. Baker, J. P. Moore, and C. D. Searles. MiR-21 is induced in endothelial cells by shear stress and modulates apoptosis and eNOS activity. Biochem. Biophys. Res. Commun. 393(4):643–648, 2010.

    Google Scholar 

  132. Wu, S. C., and Y. Zhang. Active DNA demethylation: many roads lead to Rome. Nat. Rev. Mol. Cell Biol. 11(9):607–620, 2010.

    Google Scholar 

  133. Wu, W., H. Xiao, A. Laguna-Fernandez, G. Villareal, Jr., K. C. Wang, G. G. Geary, Y. Zhang, W. C. Wang, H. D. Huang, J. Zhou, Y. S. Li, S. Chien, G. Garcia-Cardena, and J. Y. Shyy. Flow-dependent regulation of krüppel-like factor 2 is mediated by microRNA-92a. Circulation 124(5):633–641, 2011.

    Google Scholar 

  134. Yan, M. S., C. C. Matouk, and P. A. Marsden. Epigenetics of the vascular endothelium. J. Appl. Physiol. 109(3):916–926, 2010.

    Google Scholar 

  135. Yang, W. J., D. D. Yang, S. Na, G. E. Sandusky, Q. Zhang, and G. Zhao. Dicer is required for embryonic angiogenesis during mouse development. J. Biol. Chem. 280(10):9330–9335, 2005.

    Google Scholar 

  136. Zampetaki, A., L. Zeng, A. Margariti, Q. Xiao, H. Li, Z. Zhang, A. E. Pepe, G. Wang, O. Habi, E. deFalco, G. Cockerill, J. C. Mason, Y. Hu, and Q. Xu. Histone deacetylase 3 is critical in endothelial survival and atherosclerosis development in response to disturbed flow. Circulation 121(1):132–142, 2010.

    Google Scholar 

  137. Zeng, L., Q. Xiao, A. Margariti, Z. Zhang, A. Zampetaki, S. Patel, M. C. Capogrossi, Y. Hu, and Q. Xu. HDAC3 is crucial in shear- and VEGF-induced stem cell differentiation toward endothelial cells. J. Cell Biol. 174:1059–1069, 2006.

    Google Scholar 

  138. Zeng, L., Y. Zhang, S. Chien, X. Liu, and J. Y. Shyy. The role of p53 deacetylation in p21Waf1 regulation by laminar flow. J. Biol. Chem. 278(27):24594–24599, 2003.

    Google Scholar 

  139. Zernecke, A., K. Bidzhekov, H. Noels, E. Shagdarsuren, L. Gan, B. Denecke, M. Hristov, T. Koppel, M. N. Jahantigh, E. Lutgens, S. Wang, E. N. Olson, A. Schober, and C. Weber. Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci. Signal. 2(100):ra81, 2009.

    Google Scholar 

  140. Zhou, B., A. Margariti, L. Zeng, and Q. Xu. Role of histone deacetylases in vascular cell homeostasis and arteriosclerosis. Cardiovasc. Res., 2011.

  141. Zhou, J., B. O. Zhou, B. A. Lenzmeier, and J. Q. Zhou. Histone deacetylase Rpd3 antagonizes Sir2-dependent silent chromatin propagation. Nucleic Acids Res. 37(11):3699–3713, 2009.

    Google Scholar 

  142. Zhou, J., K. C. Wang, W. Wu, S. Subramaniam, J. Y. Shyy, J. J. Chiu, Y. S. Li, and S. Chien. MicroRNA-21 targets peroxisome proliferators-activated receptor-α in an autoregulatory loop to modulate flow-induced endothelial inflammation. Proc. Natl Acad. Sci. USA 108(25):10355–10360, 2011.

    Google Scholar 

  143. Zhu, N., D. Zhang, S. Chen, X. Liu, L. Lin, X. Huang, Z. Guo, J. Liu, Y. Wang, W. Yuan, and Y. Qin. Endothelial enriched microRNAs regulate angiotensin II-induced endothelial inflammation and migration. Atherosclerosis 215(2):286–293, 2011.

    Google Scholar 

Download references

Acknowledgments

This work was supported by National Science Council (Taiwan) Grants 99-2321-B-400-002 and NRPB-CV013 and National Health Research Institutes (Taiwan) Grant ME-099-PP-06. This article is a tribute to professor Shu Chien, a pioneer in the fields of bioengineering, mechanobiology, biorheology, and vascular physiology and a leading expert on how blood flow affects vessels and hence cardiovascular pathologies, for his 80th birthday.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jeng-Jiann Chiu.

Additional information

Associate Editors John Shyy and Yingxiao Wang oversaw the review of this article.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Zhou, J., Lim, S.H. & Chiu, JJ. Epigenetic Regulation of Vascular Endothelial Biology/Pathobiology and Response to Fluid Shear Stress. Cel. Mol. Bioeng. 4, 560–578 (2011). https://doi.org/10.1007/s12195-011-0199-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-011-0199-2

Keywords

Navigation