1887
Volume 2014, Issue 1
  • ISSN: 2305-7823
  • EISSN:

Abstract

The aortic valve lies in a unique hemodynamic environment, one characterized by a range of stresses (shear stress, bending forces, loading forces and strain) that vary in intensity and direction throughout the cardiac cycle. Yet, despite its changing environment, the aortic valve opens and closes over 100,000 times a day and, in the majority of human beings, will function normally over a lifespan of 70–90 years. Until relatively recently heart valves were considered passive structures that play no active role in the functioning of a valve, or in the maintenance of its integrity and durability. However, through clinical experience and basic research the aortic valve can now be characterized as a living, dynamic organ with the capacity to adapt to its complex mechanical and biomechanical environment through active and passive communication between its constituent parts. The clinical relevance of a living valve substitute in patients requiring aortic valve replacement has been confirmed. This highlights the importance of using tissue engineering to develop heart valve substitutes containing living cells which have the ability to assume the complex functioning of the native valve.

Loading

Article metrics loading...

/content/journals/10.5339/gcsp.2014.11
2014-06-01
2024-04-18
Loading full text...

Full text loading...

/deliver/fulltext/gcsp/2014/1/gcsp.2014.11.html?itemId=/content/journals/10.5339/gcsp.2014.11&mimeType=html&fmt=ahah

References

  1. El-Hamamsy I, Eryigit Z, Stevens LM, Sarang Z, George R, Clark L, Melina G, Takkenberg JJ, Yacoub MH. Long-term outcomes after autograft versus homograft aortic root replacement in adults with aortic valve disease: a randomised controlled trial. Lancet. 2010; 376:9740:524531.
    [Google Scholar]
  2. Yacoub MH, Kilner PJ, Birks EJ, Misfeld M. The aortic outflow and root: a tale of dynamism and crosstalk. Ann Thorac Surg. 1999; 68:3:S37S43.
    [Google Scholar]
  3. Lentink D, Müller UK, Stamhuis EJ, de Kat R, van Gestel W, Veldhuis LLM, Henningsson P, Hedenström A, Videler JJ, van Leeuwen JL. How swifts control their glide performance with morphing wings. Nature. 2007; 446:7139:10821085.
    [Google Scholar]
  4. Rajamannan NM. Bicuspid aortic valve disease: the role of oxidative stress in Lrp5 bone formation. Cardiovasc Pathol. 2011; 20:3:168176.
    [Google Scholar]
  5. Arjunon S, Rathan S, Jo H, Yoganathan AP. Aortic valve: mechanical environment and mechanobiology. Ann Biomed Eng. 2013; 41:7:13311346.
    [Google Scholar]
  6. Sucosky P, Padala M, Elhammali A, Balachandran K, Jo H, Yoganathan AP. Design of an ex vivo culture system to investigate the effects of shear stress on cardiovascular tissue. J Biomech Eng. 2008; 130:3:035001.
    [Google Scholar]
  7. Dagum P, Green GR, Nistal FJ, Daughters GT, Timek TA, Foppiano LE, Bolger AF, Ingels NB Jr, Miller DC. Deformational dynamics of the aortic root: modes and physiologic determinants. Circulation. 1999; 100:19:II54II62.
    [Google Scholar]
  8. Lansac E, Lim HS, Shomura Y, Lim KH, Rice NT, Goetz WA, Duran CM. Aortic root dynamics are asymmetric. J Heart Valve Dis. 2005; 14:3:400407.
    [Google Scholar]
  9. Katayama S, Umetani N, Sugiura S, Hisada T. The sinus of Valsalva relieves abnormal stress on aortic valve leaflets by facilitating smooth closure. J Thorac Cardiovasc Surg. 2008; 136:6:15281535, 35 e1.
    [Google Scholar]
  10. Robicsek F, Thubrikar MJ. Role of sinus wall compliance in aortic leaflet function. Am J Cardiol. 1999; 84:8:944946, A7.
    [Google Scholar]
  11. Thubrikar MJ, Nolan SP, Aouad J, Deck JD. Stress sharing between the sinus and leaflets of canine aortic valve. Ann Thorac Surg. 1986; 42:4:434440.
    [Google Scholar]
  12. Butcher JT, Simmons CA, Warnock JN. Mechanobiology of the aortic heart valve. J Heart Valve Dis. 2008; 17:1:6273.
    [Google Scholar]
  13. Abu-Issa R, Kirby ML. Patterning of the heart field in the chick. Dev Biol. 2008; 319:2:223233.
    [Google Scholar]
  14. Waldo KL, Hutson MR, Ward CC, Zdanowicz M, Stadt HA, Kumiski D, Abu-Issa R, Kirby ML. Secondary heart field contributes myocardium and smooth muscle to the arterial pole of the developing heart. Dev Biol. 2005; 281:1:7890.
    [Google Scholar]
  15. Moreno-Rodriguez RA, Krug EL, Reyes L, Villavicencio L, Mjaatvedt CH, Markwald RR. Bidirectional fusion of the heart-forming fields in the developing chick embryo. Dev Dyn. 2006; 235:1:191202.
    [Google Scholar]
  16. Mjaatvedt CH, Nakaoka T, Moreno-Rodriguez R, Norris RA, Kern MJ, Eisenberg CA, Turner D, Markwald RR. The outflow tract of the heart is recruited from a novel heart-forming field. Dev Biol. 2001; 238:1:97109.
    [Google Scholar]
  17. Olson EN. Gene regulatory networks in the evolution and development of the heart. Science. 2006; 313:5795:19221927.
    [Google Scholar]
  18. Dyer LA, Kirby ML. The role of secondary heart field in cardiac development. Dev Biol. 2009; 336:2:137144.
    [Google Scholar]
  19. Lucitti JL, Jones EA, Huang C, Chen J, Fraser SE, Dickinson ME. Vascular remodeling of the mouse yolk sac requires hemodynamic force. Development. 2007; 134:18:33173326.
    [Google Scholar]
  20. de la Cruz MV, Markwald RR, Krug EL, Rumenoff L, Sánchez Gómez C, Sadowinski S, Galicia TD, Gómez F, Salazar García M, Villavicencio Guzman L, Reyes Angeles L, Moreno-Rodriguez RA. Living morphogenesis of the ventricles and congenital pathology of their component parts. Cardiol Young. 2001; 11:6:588600.
    [Google Scholar]
  21. Camenisch TD, Schroeder JA, Bradley J, Klewer SE, McDonald JA. Heart-valve mesenchyme formation is dependent on hyaluronan-augmented activation of ErbB2-ErbB3 receptors. Nat Med. 2002; 8:8:850855.
    [Google Scholar]
  22. Bernanke DH, Markwald RR. Effects of hyaluronic acid on cardiac cushion tissue cells in collagen matrix cultures. Tex Rep Biol Med. 1979; 39::271285.
    [Google Scholar]
  23. Luna-Zurita L, Prados B, Grego-Bessa J, Luxán G, del Monte G, Benguría A, Adams RH, Pérez-Pomares JM, de la Pompa JL. Integration of a Notch-dependent mesenchymal gene program and Bmp2-driven cell invasiveness regulates murine cardiac valve formation. J Clin Invest. 2010; 120:10:34933507.
    [Google Scholar]
  24. Scherz PJ, Huisken J, Sahai-Hernandez P, Stainier DY. High-speed imaging of developing heart valves reveals interplay of morphogenesis and function. Development. 2008; 135:6:11791187.
    [Google Scholar]
  25. Butcher JT, Markwald RR. Valvulogenesis: the moving target. Philos Trans R Soc Lond B Biol Sci. 2007; 362:1484:14891503.
    [Google Scholar]
  26. Person AD, Klewer SE, Runyan RB. Cell biology of cardiac cushion development. Int Rev Cytol. 2005; 243::287335.
    [Google Scholar]
  27. Chang CP, Neilson JR, Bayle JH, Gestwicki JE, Kuo A, Stankunas K, Graef IA, Crabtree GR. A field of myocardial-endocardial NFAT signaling underlies heart valve morphogenesis. Cell. 2004; 118:5:649663.
    [Google Scholar]
  28. Yamagishi T, Nakajima Y, Miyazono K, Nakamura H. Bone morphogenetic protein-2 acts synergistically with transforming growth factor-beta3 during endothelial-mesenchymal transformation in the developing chick heart. J Cell Physiol. 1999; 180:1:3545.
    [Google Scholar]
  29. Mercado-Pimentel ME, Runyan RB. Multiple transforming growth factor-beta isoforms and receptors function during epithelial-mesenchymal cell transformation in the embryonic heart. Cells Tissues Organs. 2007; 185:1-3:146156.
    [Google Scholar]
  30. Holifield JS, Arlen AM, Runyan RB, Tomanek RJ. TGF-beta1, -beta2 and -beta3 cooperate to facilitate tubulogenesis in the explanted quail heart. J Vasc Res. 2004; 41:6:491498.
    [Google Scholar]
  31. Camenisch TD, Molin DG, Person A, Runyan RB, Gittenberger-de Groot AC, McDonald JA, Klewer SE. Temporal and distinct TGFbeta ligand requirements during mouse and avian endocardial cushion morphogenesis. Dev Biol. 2002; 248:1:170181.
    [Google Scholar]
  32. Boyer AS, Ayerinskas II, Vincent EB, McKinney LA, Weeks DL, Runyan RB. TGFbeta2 and TGFbeta3 have separate and sequential activities during epithelial-mesenchymal cell transformation in the embryonic heart. Dev Biol. 1999; 208:2:530545.
    [Google Scholar]
  33. Brown CB, Boyer AS, Runyan RB, Barnett JV. Requirement of type III TGF-beta receptor for endocardial cell transformation in the heart. Science. 1999; 283:5410:20802082.
    [Google Scholar]
  34. Duong TD, Erickson CA. MMP-2 plays an essential role in producing epithelial-mesenchymal transformations in the avian embryo. Dev Dyn. 2004; 229:1:4253.
    [Google Scholar]
  35. Bernanke DH, Markwald RR. Effects of two glycosaminoglycans on seeding of cardiac cushion tissue cells into a collagen-lattice culture system. Anat Rec. 1984; 210:1:2531.
    [Google Scholar]
  36. Butcher JT, Norris RA, Hoffman S, Mjaatvedt CH, Markwald RR. Periostin promotes atrioventricular mesenchyme matrix invasion and remodeling mediated by integrin signaling through Rho/PI 3-kinase. Dev Biol. 2007; 302:1:256266.
    [Google Scholar]
  37. Snider P, Hinton RB, Moreno-Rodriguez RA, Wang J, Rogers R, Lindsley A, Li F, Ingram DA, Menick D, Field L, Firulli AB, Molkentin JD, Markwald R, Conway SJ. Periostin is required for maturation and extracellular matrix stabilization of noncardiomyocyte lineages of the heart. Circ Res. 2008; 102:7:752760.
    [Google Scholar]
  38. Zhou B, Wu B, Tompkins KL, Boyer KL, Grindley JC, Baldwin HS. Characterization of Nfatc1 regulation identifies an enhancer required for gene expression that is specific to pro-valve endocardial cells in the developing heart. Development. 2005; 132:5:11371146.
    [Google Scholar]
  39. Forouhar AS, Liebling M, Hickerson A, Nasiraei-Moghaddam A, Tsai HJ, Hove JR, Fraser SE, Dickinson ME, Gharib M. The embryonic vertebrate heart tube is a dynamic suction pump. Science. 2006; 312:5774:751753.
    [Google Scholar]
  40. Butcher JT, McQuinn TC, Sedmera D, Turner D, Markwald RR. Transitions in early embryonic atrioventricular valvular function correspond with changes in cushion biomechanics that are predictable by tissue composition. Circ Res. 2007; 100:10:15031511.
    [Google Scholar]
  41. Sugi Y, Ito N, Szebenyi G, Myers K, Fallon JF, Mikawa T, Markwald RR. Fibroblast growth factor (FGF)-4 can induce proliferation of cardiac cushion mesenchymal cells during early valve leaflet formation. Dev Biol. 2003; 258:2:252263.
    [Google Scholar]
  42. Yalcin HC, Shekhar A, McQuinn TC, Butcher JT. Hemodynamic patterning of the avian atrioventricular valve. Dev Dyn. 2011; 240:1:2335.
    [Google Scholar]
  43. Snarr BS, Wirrig EE, Phelps AL, Trusk TC, Wessels A. A spatiotemporal evaluation of the contribution of the dorsal mesenchymal protrusion to cardiac development. Dev Dyn. 2007; 236:5:12871294.
    [Google Scholar]
  44. Webb S, Qayyum SR, Anderson RH, Lamers WH, Richardson MK. Septation and separation within the outflow tract of the developing heart. J Anat. 2003; 202:4:327342.
    [Google Scholar]
  45. Jain R, Engleka KA, Rentschler SL, Manderfield LJ, Li L, Yuan L, Epstein JA. Cardiac neural crest orchestrates remodeling and functional maturation of mouse semilunar valves. J Clin Invest. 2011; 121:1:422430.
    [Google Scholar]
  46. Kirby ML, Gale TF, Stewart DE. Neural crest cells contribute to normal aorticopulmonary septation. Science. 1983; 220:4601:10591061.
    [Google Scholar]
  47. Qayyum SR, Webb S, Anderson RH, Verbeek FJ, Brown NA, Richardson MK. Septation and valvar formation in the outflow tract of the embryonic chick heart. Anat Rec. 2001; 264:3:273283.
    [Google Scholar]
  48. van den Hoff MJ, Moorman AF, Ruijter JM, Lamers WH, Bennington RW, Markwald RR, Wessels A. Myocardialization of the cardiac outflow tract. Dev Biol. 1999; 212:2:477490.
    [Google Scholar]
  49. Lincoln J, Alfieri CM, Yutzey KE. Development of heart valve leaflets and supporting apparatus in chicken and mouse embryos. Dev Dyn. 2004; 230:2:239250.
    [Google Scholar]
  50. Ranger AM, Grusby MJ, Hodge MR, Gravallese EM, de la Brousse FC, Hoey T, Mickanin C, Baldwin HS, Glimcher LH. The transcription factor NF-ATc is essential for cardiac valve formation. Nature. 1998; 392:6672:186190.
    [Google Scholar]
  51. Jackson LF, Qiu TH, Sunnarborg SW, Chang A, Zhang C, Patterson C, Lee DC. Defective valvulogenesis in HB-EGF and TACE-null mice is associated with aberrant BMP signaling. EMBO J. 2003; 22:11:27042716.
    [Google Scholar]
  52. de Lange FJ, Moorman AF, Anderson RH, Männer J, Soufan AT, de Gier-de Vries C, Schneider MD, Webb S, van den Hoff MJ, Christoffels VM. Lineage and morphogenetic analysis of the cardiac valves. Circ Res. 2004; 95:6:645654.
    [Google Scholar]
  53. Wessels A, van den Hoff MJ, Adamo RF, Phelps AL, Lockhart MM, Sauls K, Briggs LE, Norris RA, van Wijk B, Perez-Pomares JM, Dettman RW, Burch JB. Epicardially derived fibroblasts preferentially contribute to the parietal leaflets of the atrioventricular valves in the murine heart. Dev Biol. 2012; 366:2:111124.
    [Google Scholar]
  54. Colvee E, Hurle JM. Maturation of the extracellular material of the semilunar heart values in the mouse. A histochemical analysis of collagen and mucopolysaccharides. Anat Embryol (Berl). 1981; 162:3:343352.
    [Google Scholar]
  55. Armstrong EJ, Bischoff J. Heart valve development: endothelial cell signaling and differentiation. Circ Res. 2004; 95:5:459470.
    [Google Scholar]
  56. Chiu YN, Norris RA, Mahler G, Recknagel A, Butcher JT. Transforming growth factor beta, bone morphogenetic protein, and vascular endothelial growth factor mediate phenotype maturation and tissue remodeling by embryonic valve progenitor cells: relevance for heart valve tissue engineering. Tissue Eng Part A. 2010; 16:11:33753383.
    [Google Scholar]
  57. Chakraborty S, Combs MD, Yutzey KE. Transcriptional regulation of heart valve progenitor cells. Pediatr Cardiol. 2010; 31:3:414421.
    [Google Scholar]
  58. Galvin KM, Donovan MJ, Lynch CA, Meyer RI, Paul RJ, Lorenz JN, Fairchild-Huntress V, Dixon KL, Dunmore JH, Gimbrone MA Jr, Falb D, Huszar D. A role for smad6 in development and homeostasis of the cardiovascular system. Nat Genet. 2000; 24:2:171174.
    [Google Scholar]
  59. Peacock JD, Levay AK, Gillaspie DB, Tao G, Lincoln J. Reduced sox9 function promotes heart valve calcification phenotypes in vivo. Circ Res. 2010; 106:4:712719.
    [Google Scholar]
  60. Levay AK, Peacock JD, Lu Y, Koch M, Hinton RB Jr, Kadler KE, Lincoln J. Scleraxis is required for cell lineage differentiation and extracellular matrix remodeling during murine heart valve formation in vivo. Circ Res. 2008; 103:9:948956.
    [Google Scholar]
  61. Lee MP, Yutzey KE. Twist1 directly regulates genes that promote cell proliferation and migration in developing heart valves. PLoS One. 2011; 6:12:e29758.
    [Google Scholar]
  62. Lincoln J, Alfieri CM, Yutzey KE. BMP and FGF regulatory pathways control cell lineage diversification of heart valve precursor cells. Dev Biol. 2006; 292:2:292302.
    [Google Scholar]
  63. Cheek JD, Wirrig EE, Alfieri CM, James JF, Yutzey KE. Differential activation of valvulogenic, chondrogenic, and osteogenic pathways in mouse models of myxomatous and calcific aortic valve disease. J Mol Cell Cardiol. 2012; 52:3:689700.
    [Google Scholar]
  64. Manner J. Cardiac looping in the chick embryo: a morphological review with special reference to terminological and biomechanical aspects of the looping process. Anat Rec. 2000; 259:3:248262.
    [Google Scholar]
  65. Hove JR, Koster RW, Forouhar AS, Acevedo-Bolton G, Fraser SE, Gharib M. Intracardiac fluid forces are an essential epigenetic factor for embryonic cardiogenesis. Nature. 2003; 421:6919:172177.
    [Google Scholar]
  66. Vermot J, Forouhar AS, Liebling M, Wu D, Plummer D, Gharib M, Fraser SE. Reversing blood flows act through klf2a to ensure normal valvulogenesis in the developing heart. PLoS Biol. 2009; 7:11:e1000246.
    [Google Scholar]
  67. Egorova AD, Khedoe PP, Goumans MJ, Yoder BK, Nauli SM, ten Dijke P, Poelmann RE, Hierck BP. Lack of primary cilia primes shear-induced endothelial-to-mesenchymal transition. Circ Res. 2011; 108:9:10931101.
    [Google Scholar]
  68. Buskohl PR, Jenkins JT, Butcher JT. Computational simulation of hemodynamic-driven growth and remodeling of embryonic atrioventricular valves. Biomech Model Mechanobiol. 2012; 11:8:12051217.
    [Google Scholar]
  69. Biechler SV, Potts JD, Yost MJ, Junor L, Goodwin RL, Weidner JW. Mathematical modeling of flow-generated forces in an in vitro system of cardiac valve development. Ann Biomed Eng. 2010; 38:1:109117.
    [Google Scholar]
  70. Goodwin RL, Nesbitt T, Price RL, Wells JC, Yost MJ, Potts JD. Three-dimensional model system of valvulogenesis. Dev Dyn. 2005; 233:1:122129.
    [Google Scholar]
  71. Tan H, Biechler S, Junor L, Yost MJ, Dean D, Li J, Potts JD, Goodwin RL. Fluid flow forces and rhoA regulate fibrous development of the atrioventricular valves. Dev Biol. 2013; 374:2:345356.
    [Google Scholar]
  72. Hu N, Christensen DA, Agrawal AK, Beaumont C, Clark EB, Hawkins JA. Dependence of aortic arch morphogenesis on intracardiac blood flow in the left atrial ligated chick embryo. Anat Rec (Hoboken). 2009; 292:5:652660.
    [Google Scholar]
  73. Reckova M, Rosengarten C, deAlmeida A, Stanley CP, Wessels A, Gourdie RG, Thompson RP, Sedmera D. Hemodynamics is a key epigenetic factor in development of the cardiac conduction system. Circ Res. 2003; 93:1:7785.
    [Google Scholar]
  74. Sedmera D, Hu N, Weiss KM, Keller BB, Denslow S, Thompson RP. Cellular changes in experimental left heart hypoplasia. Anat Rec. 2002; 267:2:137145.
    [Google Scholar]
  75. Sedmera D, Pexieder T, Rychterova V, Hu N, Clark EB. Remodeling of chick embryonic ventricular myoarchitecture under experimentally changed loading conditions. Anat Rec. 1999; 254:2:238252.
    [Google Scholar]
  76. Yalcin HC, Shekhar A, Nishimura N, Rane AA, Schaffer CB, Butcher JT. Two-photon microscopy-guided femtosecond-laser photoablation of avian cardiogenesis: noninvasive creation of localized heart defects. Am J Physiol Heart Circ Physiol. 2010; 299:5:H1728H1735.
    [Google Scholar]
  77. Yano K, Gale D, Massberg S, Cheruvu PK, Monahan-Earley R, Morgan ES, Haig D, von Andrian UH, Dvorak AM, Aird WC. Phenotypic heterogeneity is an evolutionarily conserved feature of the endothelium. Blood. 2007; 109:2:613615.
    [Google Scholar]
  78. Otto CM, Kuusisto J, Reichenbach DD, Gown AM, O'Brien KD. Characterization of the early lesion of ’degenerative’ valvular aortic stenosis. Histological and immunohistochemical studies. Circulation. 1994; 90:2:844853.
    [Google Scholar]
  79. Imberti B, Seliktar D, Nerem RM, Remuzzi A. The response of endothelial cells to fluid shear stress using a co-culture model of the arterial wall. Endothelium. 2002; 9:1:1123.
    [Google Scholar]
  80. Passerini AG, Polacek DC, Shi C, Francesco NM, Manduchi E, Grant GR, Pritchard WF, Powell S, Chang GY, Stoeckert CJ Jr, Davies PF. Coexisting proinflammatory and antioxidative endothelial transcription profiles in a disturbed flow region of the adult porcine aorta. Proc Natl Acad Sci USA. 2004; 101:8:24822487.
    [Google Scholar]
  81. Butcher JT, Penrod AM, Garcia AJ, Nerem RM. Unique morphology and focal adhesion development of valvular endothelial cells in static and fluid flow environments. Arterioscler Thromb Vasc Biol. 2004; 24:8:14291434.
    [Google Scholar]
  82. Deck JD. Endothelial cell orientation on aortic valve leaflets. Cardiovasc Res. 1986; 20:10:760767.
    [Google Scholar]
  83. Farivar RS, Cohn LH, Soltesz EG, Mihaljevic T, Rawn JD, Byrne JG. Transcriptional profiling and growth kinetics of endothelium reveals differences between cells derived from porcine aorta versus aortic valve. Eur J Cardiothorac Surg. 2003; 24:4:527534.
    [Google Scholar]
  84. Davies PF. Flow-mediated endothelial mechanotransduction. Physiol Rev. 1995; 75:3:519560.
    [Google Scholar]
  85. Lopez-Quintero SV, Amaya R, Pahakis M, Tarbell JM. The endothelial glycocalyx mediates shear-induced changes in hydraulic conductivity. Am J Physiol Heart Circ Physiol. 2009; 296:5:H1451H1456.
    [Google Scholar]
  86. Pahakis MY, Kosky JR, Dull RO, Tarbell JM. The role of endothelial glycocalyx components in mechanotransduction of fluid shear stress. Biochem Biophys Res Commun. 2007; 355:1:228233.
    [Google Scholar]
  87. Wang N, Butler JP, Ingber DE. Mechanotransduction across the cell surface and through the cytoskeleton. Science. 1993; 260:5111:11241127.
    [Google Scholar]
  88. Ku DD, Nelson JM, Caulfield JB, Winn MJ. Release of endothelium-derived relaxing factors from canine cardiac valves. J Cardiovasc Pharmacol. 1990; 16:2:212218.
    [Google Scholar]
  89. Misfeld M, Morrison K, Sievers H, Yacoub MH, Chester AH. Localization of immunoreactive endothelin and characterization of its receptors in aortic cusps. J Heart Valve Dis. 2002; 11:4:472476 discussion 6-7.
    [Google Scholar]
  90. Pompilio G, Rossoni G, Sala A, Polvani GL, Berti F, Dainese L, Porqueddu M, Biglioli P. Endothelial-dependent dynamic and antithrombotic properties of porcine aortic and pulmonary valves. Ann Thorac Surg. 1998; 65:4:986992.
    [Google Scholar]
  91. Caro CG. Discovery of the role of wall shear in atherosclerosis. Arterioscler Thromb Vasc Biol. 2009; 29:2:158161.
    [Google Scholar]
  92. Chatzizisis YS, Coskun AU, Jonas M, Edelman ER, Feldman CL, Stone PH. Role of endothelial shear stress in the natural history of coronary atherosclerosis and vascular remodeling: molecular, cellular, and vascular behavior. J Am Coll Cardiol. 2007; 49:25:23792393.
    [Google Scholar]
  93. Butcher JT, Tressel S, Johnson T, Turner D, Sorescu G, Jo H, Nerem RM. Transcriptional profiles of valvular and vascular endothelial cells reveal phenotypic differences: influence of shear stress. Arterioscler Thromb Vasc Biol. 2006; 26:1:6977.
    [Google Scholar]
  94. Rabkin E, Aikawa M, Stone JR, Fukumoto Y, Libby P, Schoen FJ. Activated interstitial myofibroblasts express catabolic enzymes and mediate matrix remodeling in myxomatous heart valves. Circulation. 2001; 104:21:25252532.
    [Google Scholar]
  95. Walker GA, Masters KS, Shah DN, Anseth KS, Leinwand LA. Valvular myofibroblast activation by transforming growth factor-beta: implications for pathological extracellular matrix remodeling in heart valve disease. Circ Res. 2004; 95:3:253260.
    [Google Scholar]
  96. Taylor PM, Allen SP, Yacoub MH. Phenotypic and functional characterization of interstitial cells from human heart valves, pericardium and skin. J Heart Valve Dis. 2000; 9:1:150158.
    [Google Scholar]
  97. Brand NJ, Roy A, Hoare G, Chester A, Yacoub MH. Cultured interstitial cells from human heart valves express both specific skeletal muscle and non-muscle markers. Int J Biochem Cell Biol. 2006; 38:1:3042.
    [Google Scholar]
  98. Rabkin-Aikawa E, Farber M, Aikawa M, Schoen FJ. Dynamic and reversible changes of interstitial cell phenotype during remodeling of cardiac valves. J Heart Valve Dis. 2004; 13:5:841847.
    [Google Scholar]
  99. Liu AC, Joag VR, Gotlieb AI. The emerging role of valve interstitial cell phenotypes in regulating heart valve pathobiology. Am J Pathol. 2007; 171:5:14071418.
    [Google Scholar]
  100. Osman L, Yacoub MH, Latif N, Amrani M, Chester AH. Role of human valve interstitial cells in valve calcification and their response to atorvastatin. Circulation. 2006; 114:1:I547I552.
    [Google Scholar]
  101. Merryman WD, Youn I, Lukoff HD, Krueger PM, Guilak F, Hopkins RA, Sacks MS. Correlation between heart valve interstitial cell stiffness and transvalvular pressure: implications for collagen biosynthesis. Am J Physiol Heart Circ Physiol. 2006; 290:1:H224H231.
    [Google Scholar]
  102. Latif N, Sarathchandra P, Taylor PM, Antoniw J, Brand N, Yacoub MH. Characterization of molecules mediating cell-cell communication in human cardiac valve interstitial cells. Cell Biochem Biophys. 2006; 45:3:255264.
    [Google Scholar]
  103. Latif N, Sarathchandra P, Thomas PS, Antoniw J, Batten P, Chester AH, Taylor PM, Yacoub MH. Characterization of structural and signaling molecules by human valve interstitial cells and comparison to human mesenchymal stem cells. J Heart Valve Dis. 2007; 16:1:5666.
    [Google Scholar]
  104. Balachandran K, Sucosky P, Jo H, Yoganathan AP. Elevated cyclic stretch alters matrix remodeling in aortic valve cusps: implications for degenerative aortic valve disease. Am J Physiol Heart Circ Physiol. 2009; 296:3:H756H764.
    [Google Scholar]
  105. Ku CH, Johnson PH, Batten P, Sarathchandra P, Chambers RC, Taylor PM, Yacoub MH, Chester AH. Collagen synthesis by mesenchymal stem cells and aortic valve interstitial cells in response to mechanical stretch. Cardiovasc Res. 2006; 71:3:548556.
    [Google Scholar]
  106. Merryman WD, Lukoff HD, Long RA, Engelmayr GC Jr, Hopkins RA, Sacks MS. Synergistic effects of cyclic tension and transforming growth factor-beta1 on the aortic valve myofibroblast. Cardiovasc Pathol. 2007; 16:5:268276.
    [Google Scholar]
  107. Schoen FJ. Evolving concepts of cardiac valve dynamics: the continuum of development, functional structure, pathobiology, and tissue engineering. Circulation. 2008; 118:18:18641880.
    [Google Scholar]
  108. Latif N, Sarathchandra P, Taylor PM, Antoniw J, Yacoub MH. Molecules mediating cell-ECM and cell-cell communication in human heart valves. Cell Biochem Biophys. 2005; 43:2:275287.
    [Google Scholar]
  109. Blaschuk KL, Frost EE, ffrench-Constant C. The regulation of proliferation and differentiation in oligodendrocyte progenitor cells by alphaV integrins. Development. 2000; 127:9:19611969.
    [Google Scholar]
  110. Mawatari K, Liu B, Kent KC. Activation of integrin receptors is required for growth factor-induced smooth muscle cell dysfunction. J Vasc Surg. 2000; 31:2:375381.
    [Google Scholar]
  111. Tsuji T, Waga I, Tezuka K, Kamada M, Yatsunami K, Kodama H. Integrin beta2 (CD18)-mediated cell proliferation of HEL cells on a hematopoietic-supportive bone marrow stromal cell line, HESS-5 cells. Blood. 1998; 91:4:12631271.
    [Google Scholar]
  112. El-Hamamsy I, Yacoub MH, Chester AH. Neuronal regulation of aortic valve cusps. Curr Vasc Pharmacol. 2009; 7:1:4046.
    [Google Scholar]
  113. Marron K, Yacoub MH, Polak JM, Sheppard MN, Fagan D, Whitehead BF, de Leval MR, Anderson RH, Wharton J. Innervation of human atrioventricular and arterial valves. Circulation. 1996; 94:3:368375.
    [Google Scholar]
  114. Kawano H, Kawai S, Shirai T, Okada R. Morphological study on vagal innervation in human atrioventricular valves using histochemical method. Jpn Circ J. 1993; 57:8:753759.
    [Google Scholar]
  115. Kawano H, Shirai T, Kawano Y, Okada R. Morphological study of vagal innervation in human semilunar valves using a histochemical method. Jpn Circ J. 1996; 60:1:6266.
    [Google Scholar]
  116. De Biasi S, Vitellaro-Zuccarello L. Intrinsic innervation of porcine semilunar heart valves. Anat Embryol (Berl). 1982; 165:1:7179.
    [Google Scholar]
  117. Steele PA, Gibbins IL, Morris JL. Projections of intrinsic cardiac neurons to different targets in the guinea-pig heart. J Auton Nerv Syst. 1996; 56:3:191200.
    [Google Scholar]
  118. Chester AH, Kershaw JD, Sarathchandra P, Yacoub MH. Localisation and function of nerves in the aortic root. J Mol Cell Cardiol. 2008; 44:6:10451052.
    [Google Scholar]
  119. Billiar KL, Sacks MS. Biaxial mechanical properties of the native and glutaraldehyde-treated aortic valve cusp: Part II–A structural constitutive model. J Biomech Eng. 2000; 122:4:327335.
    [Google Scholar]
  120. Billiar KL, Sacks MS. Biaxial mechanical properties of the natural and glutaraldehyde treated aortic valve cusp–Part I: Experimental results. J Biomech Eng. 2000; 122:1:2330.
    [Google Scholar]
  121. Stella JA, Liao J, Sacks MS. Time-dependent biaxial mechanical behavior of the aortic heart valve leaflet. J Biomech. 2007; 40:14:31693177.
    [Google Scholar]
  122. Butcher JT, Nerem RM. Valvular endothelial cells regulate the phenotype of interstitial cells in co-culture: effects of steady shear stress. Tissue Eng. 2006; 12:4:905915.
    [Google Scholar]
  123. El-Hamamsy I, Balachandran K, Yacoub MH, Stevens LM, Sarathchandra P, Taylor PM, Yoganathan AP, Chester AH. Endothelium-dependent regulation of the mechanical properties of aortic valve cusps. J Am Coll Cardiol. 2009; 53:16:14481455.
    [Google Scholar]
  124. Aicher D, Urbich C, Zeiher A, Dimmeler S, Schafers HJ. Endothelial nitric oxide synthase in bicuspid aortic valve disease. Ann Thorac Surg. 2007; 83:4:12901294.
    [Google Scholar]
  125. Lee TC, Zhao YD, Courtman DW, Stewart DJ. Abnormal aortic valve development in mice lacking endothelial nitric oxide synthase. Circulation. 2000; 101:20:23452348.
    [Google Scholar]
  126. Davies JE, Parker KH, Francis DP, Hughes AD, Mayet J. What is the role of the aorta in directing coronary blood flow? Heart. 2008; 94:12:15451547.
    [Google Scholar]
  127. Grande-Allen KJ, Cochran RP, Reinhall PG, Kunzelman KS. Re-creation of sinuses is important for sparing the aortic valve: a finite element study. J Thorac Cardiovasc Surg. 2000; 119:4 Pt 1:753763.
    [Google Scholar]
  128. El-Hamamsy I, Yacoub MH. A measured approach to managing the aortic root in patients with bicuspid aortic valve disease. Curr Cardiol Rep. 2009; 11:2:94100.
    [Google Scholar]
  129. Grande KJ, Cochran RP, Reinhall PG, Kunzelman KS. Mechanisms of aortic valve incompetence: finite element modeling of aortic root dilatation. Ann Thorac Surg. 2000; 69:6:18511857.
    [Google Scholar]
  130. Owens DS, Otto CM. Is it time for a new paradigm in calcific aortic valve disease? JACC Cardiovasc Imaging. 2009; 2:8:928930.
    [Google Scholar]
  131. Thubrikar MJ, Aouad J, Nolan SP. Patterns of calcific deposits in operatively excised stenotic or purely regurgitant aortic valves and their relation to mechanical stress. Am J Cardiol. 1986; 58:3:304308.
    [Google Scholar]
  132. Ingber DE. The mechanochemical basis of cell and tissue regulation. Mech Chem Biosyst. 2004; 1:1:5368.
    [Google Scholar]
  133. Van der Heiden K, Groenendijk BC, Hierck BP, Hogers B, Koerten HK, Mommaas AM, Gittenberger-de Groot AC, Poelmann RE. Monocilia on chicken embryonic endocardium in low shear stress areas. Dev Dyn. 2006; 235:1:1928.
    [Google Scholar]
  134. Dumbauld DW, Michael KE, Hanks SK, Garcia AJ. Focal adhesion kinase-dependent regulation of adhesive forces involves vinculin recruitment to focal adhesions. Biol Cell. 2010; 102:4:203213.
    [Google Scholar]
  135. Xu Y, Bismar TA, Su J, Xu B, Kristiansen G, Varga Z, Teng L, Ingber DE, Mammoto A, Kumar R, Alaoui-Jamali MA. Filamin A regulates focal adhesion disassembly and suppresses breast cancer cell migration and invasion. J Exp Med. 2010; 207:11:24212437.
    [Google Scholar]
  136. Kyndt F, Gueffet JP, Probst V, Jaafar P, Legendre A, Le Bouffant F, Toquet C, Roy E, McGregor L, Lynch SA, Newbury-Ecob R, Tran V, Young I, Trochu JN, Le Marec H, Schott JJ. Mutations in the gene encoding filamin A as a cause for familial cardiac valvular dystrophy. Circulation. 2007; 115:1:4049.
    [Google Scholar]
  137. Gu X, Masters KS. Role of the Rho pathway in regulating valvular interstitial cell phenotype and nodule formation. Am J Physiol Heart Circ Physiol. 2011; 300:2:H448H458.
    [Google Scholar]
  138. Helmke BP, Davies PF. The cytoskeleton under external fluid mechanical forces: hemodynamic forces acting on the endothelium. Ann Biomed Eng. 2002; 30:3:284296.
    [Google Scholar]
  139. Ingber DE. Tensegrity: the architectural basis of cellular mechanotransduction. Annu Rev Physiol. 1997; 59::575599.
    [Google Scholar]
  140. Wang N, Tytell JD, Ingber DE. Mechanotransduction at a distance: mechanically coupling the extracellular matrix with the nucleus. Nat Rev Mol Cell Biol. 2009; 10:1:7582.
    [Google Scholar]
  141. Lammerding J. Mechanics of the nucleus. Compr Physiol. 2011; 1:2:783807.
    [Google Scholar]
  142. Gould RA, Sinha R, Aziz H, Rouf R, Dietz HC 3rd, Judge DP, Butcher J. Multi-scale biomechanical remodeling in aging and genetic mutant murine mitral valve leaflets: insights into Marfan syndrome. PLoS One. 2012; 7:9:e44639.
    [Google Scholar]
  143. Weiler M, Yap CH, Balachandran K, Padala M, Yoganathan AP. Regional analysis of dynamic deformation characteristics of native aortic valve leaflets. J Biomech. 2011; 44:8:14591465.
    [Google Scholar]
  144. Gould RA, Chin K, Santisakultarm TP, Dropkin A, Richards JM, Schaffer CB, Butcher JT. Cyclic strain anisotropy regulates valvular interstitial cell phenotype and tissue remodeling in three-dimensional culture. Acta Biomater. 2012; 8:5:17101719.
    [Google Scholar]
  145. Kural MH, Billiar KL. Mechanoregulation of valvular interstitial cell phenotype in the third dimension. Biomaterials. 2014; 35:4:11281137.
    [Google Scholar]
  146. Holliday CJ, Ankeny RF, Jo H, Nerem RM. Discovery of shear- and side-specific mRNAs and miRNAs in human aortic valvular endothelial cells. Am J Physiol Heart Circ Physiol. 2011; 301:3:H856H867.
    [Google Scholar]
  147. Metzler SA, Digesu CS, Howard JI, Filip To SD, Warnock JN. Live en face imaging of aortic valve leaflets under mechanical stress. Biomech Model Mechanobiol. 2012; 11:3-4:355361.
    [Google Scholar]
  148. Clark-Greuel JN, Connolly JM, Sorichillo E, Narula NR, Rapoport HS, Mohler ER 3rd, Gorman JH 3rd, Gorman RC, Levy RJ. Transforming growth factor-beta1 mechanisms in aortic valve calcification: increased alkaline phosphatase and related events. Ann Thorac Surg. 2007; 83:3:946953.
    [Google Scholar]
  149. Mohler ER 3rd, Chawla MK, Chang AW, Vyavahare N, Levy RJ, Graham L, Gannon FH. Identification and characterization of calcifying valve cells from human and canine aortic valves. J Heart Valve Dis. 1999; 8:3:254260.
    [Google Scholar]
  150. Ngo DT, Sverdlov AL, Willoughby SR, Nightingale AK, Chirkov YY, McNeil JJ, Horowitz JD. Determinants of occurrence of aortic sclerosis in an aging population. JACC Cardiovasc Imaging. 2009; 2:8:919927.
    [Google Scholar]
  151. Miller JD, Chu Y, Brooks RM, Richenbacher WE, Pena-Silva R, Heistad DD. Dysregulation of antioxidant mechanisms contributes to increased oxidative stress in calcific aortic valvular stenosis in humans. J Am Coll Cardiol. 2008; 52:10:843850.
    [Google Scholar]
  152. Ferdous Z, Jo H, Nerem RM. Strain magnitude-dependent calcific marker expression in valvular and vascular cells. Cells Tissues Organs. 2013; 197:5:372383.
    [Google Scholar]
  153. Mohler ER, Gannon F, Reynolds C, Zimmerman R, Keane MG, Kaplan FS. Bone formation and inflammation in cardiac valves. Circulation. 2001; 103:11:15221528.
    [Google Scholar]
  154. Yang X, Meng X, Su X, Mauchley DC, Ao L, Cleveland JC Jr, Fullerton DA. Bone morphogenic protein 2 induces Runx2 and osteopontin expression in human aortic valve interstitial cells: role of Smad1 and extracellular signal-regulated kinase 1/2. J Thorac Cardiovasc Surg. 2009; 138:4:10081015.
    [Google Scholar]
  155. Yip CY, Chen JH, Zhao R, Simmons CA. Calcification by valve interstitial cells is regulated by the stiffness of the extracellular matrix. Arterioscler Thromb Vasc Biol. 2009; 29:6:936942.
    [Google Scholar]
  156. Balachandran K, Sucosky P, Jo H, Yoganathan AP. Elevated cyclic stretch induces aortic valve calcification in a bone morphogenic protein-dependent manner. Am J Pathol. 2010; 177:1:4957.
    [Google Scholar]
  157. Chen JH, Yip CY, Sone ED, Simmons CA. Identification and characterization of aortic valve mesenchymal progenitor cells with robust osteogenic calcification potential. Am J Pathol. 2009; 174:3:11091119.
    [Google Scholar]
  158. Richards J, El-Hamamsy I, Chen S, Sarang Z, Sarathchandra P, Yacoub MH, Chester AH, Butcher JT. Side-specific endothelial-dependent regulation of aortic valve calcification: interplay of hemodynamics and nitric oxide signaling. Am J Pathol. 2013; 182:5:19221931.
    [Google Scholar]
  159. Kennedy JA, Hua X, Mishra K, Murphy GA, Rosenkranz AC, Horowitz JD. Inhibition of calcifying nodule formation in cultured porcine aortic valve cells by nitric oxide donors. Eur J Pharmacol. 2009; 602:1:2835.
    [Google Scholar]
  160. Simmons CA, Zilberberg J, Davies PF. A rapid, reliable method to isolate high quality endothelial RNA from small spatially-defined locations. Ann Biomed Eng. 2004; 32:10:14531459.
    [Google Scholar]
  161. Simmons CA, Grant GR, Manduchi E, Davies PF. Spatial heterogeneity of endothelial phenotypes correlates with side-specific vulnerability to calcification in normal porcine aortic valves. Circ Res. 2005; 96:7:792799.
    [Google Scholar]
  162. Peltonen TO, Taskinen P, Soini Y, Rysa J, Ronkainen J, Ohtonen P, Satta J, Juvonen T, Ruskoaho H, Leskinen H. Distinct downregulation of C-type natriuretic peptide system in human aortic valve stenosis. Circulation. 2007; 116:11:12831289.
    [Google Scholar]
  163. Yip CY, Blaser MC, Mirzaei Z, Zhong X, Simmons CA. Inhibition of pathological differentiation of valvular interstitial cells by C-type natriuretic peptide. Arterioscler Thromb Vasc Biol. 2011; 31:8:18811889.
    [Google Scholar]
  164. Chester AH. Molecular and cellular mechanisms of valve calcification. Aswan Heart Cent Sci Pract Ser. 2011; 4::19.
    [Google Scholar]
  165. Helderman F, Segers D, de Crom R, Hierck BP, Poelmann RE, Evans PC, Krams R. Effect of shear stress on vascular inflammation and plaque development. Curr Opin Lipidol. 2007; 18:5:527533.
    [Google Scholar]
  166. Partridge J, Carlsen H, Enesa K, Chaudhury H, Zakkar M, Luong L, Kinderlerer A, Johns M, Blomhoff R, Mason JC, Haskard DO, Evans PC. Laminar shear stress acts as a switch to regulate divergent functions of NF-kappaB in endothelial cells. FASEB J. 2007; 21:13:35533561.
    [Google Scholar]
  167. Steinmetz M, Skowasch D, Wernert N, Welsch U, Preusse CJ, Welz A, Nickenig G, Bauriedel G. Differential profile of the OPG/RANKL/RANK-system in degenerative aortic native and bioprosthetic valves. J Heart Valve Dis. 2008; 17:2:187193.
    [Google Scholar]
  168. Jian B, Narula N, Li QY, Mohler ER 3rd, Levy RJ. Progression of aortic valve stenosis: TGF-beta1 is present in calcified aortic valve cusps and promotes aortic valve interstitial cell calcification via apoptosis. Ann Thorac Surg. 2003; 75:2:457465, ; discussion 65-6.
    [Google Scholar]
  169. Hakuno D, Kimura N, Yoshioka M, Fukuda K. Molecular mechanisms underlying the onset of degenerative aortic valve disease. J Mol Med (Berl). 2009; 87:1:1724.
    [Google Scholar]
  170. Latif N, Sarathchandra P, Chester AH, Yacoub MH. Expression of smooth muscle cell markers and co-activators in calcified aortic valves. Eur Heart J. 2014;, In Press.
    [Google Scholar]
  171. Torii R, El-Hamamsy I, Donya M, Babu-Narayan SV, Ibrahim M, Kilner PJ, Mohiaddin RH, Xu XY, Yacoub MH. Integrated morphologic and functional assessment of the aortic root after different tissue valve root replacement procedures. J Thorac Cardiovasc Surg. 2012; 143:6:14221428.
    [Google Scholar]
  172. Aikawa E, Nahrendorf M, Sosnovik D, Lok VM, Jaffer FA, Aikawa M, Weissleder R. Multimodality molecular imaging identifies proteolytic and osteogenic activities in early aortic valve disease. Circulation. 2007; 115:3:377386.
    [Google Scholar]
  173. Bertazzo S, Gentleman E, Cloyd KL, Chester AH, Yacoub MH, Stevens MM. Nano-analytical electron microscopy reveals fundamental insights into human cardiovascular tissue calcification. Nat Mater. 2013; 12:6:576583.
    [Google Scholar]
  174. Yacoub MH, Takkenberg JJ. Will heart valve tissue engineering change the world? Nat Clin Pract Cardiovasc Med. 2005; 2:2:6061.
    [Google Scholar]
  175. Lansac E, Lim HS, Shomura Y, Lim KH, Rice NT, Goetz W, Acar C, Duran CM. A four-dimensional study of the aortic root dynamics. Eur J Cardiothorac Surg. 2002; 22:4:497503.
    [Google Scholar]
  176. High FA, Epstein JA. The multifaceted role of Notch in cardiac development and disease. Nat Rev Genet. 2008; 9:1:4961.
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journals/10.5339/gcsp.2014.11
Loading
/content/journals/10.5339/gcsp.2014.11
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error