Comprehensive Physiology Wiley Online Library

Pathophysiology of Aortic Stenosis and Mitral Regurgitation

Full Article on Wiley Online Library



ABSTRACT

The global impact of the spectrum of valve diseases is a crucial, fast‐growing, and underrecognized health problem. The most prevalent valve diseases, requiring surgical intervention, are represented by calcific and degenerative processes occurring in heart valves, in particular, aortic and mitral valve. Due to the increasing elderly population, these pathologies will gain weight in the global health burden. The two most common valve diseases are aortic valve stenosis (AVS) and mitral valve regurgitation (MR). AVS is the most commonly encountered valve disease nowadays and affects almost 5% of elderly population. In particular, AVS poses a great challenge due to the multiple comorbidities and frailty of this patient subset. MR is also a common valve pathology and has an estimated prevalence of 3% in the general population, affecting more than 176 million people worldwide. This review will focus on pathophysiological changes in both these valve diseases, starting from the description of the anatomical aspects of normal valve, highlighting all the main cellular and molecular features involved in the pathological progression and cardiac consequences. This review also evaluates the main approaches in clinical management of these valve diseases, taking into account of the main published clinical guidelines. © 2017 American Physiological Society. Compr Physiol 7:799‐818, 2017.

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

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Aortic valve anatomy. Structure and nomenclature of aortic root components. The upper side of the figure represents thoracic aortic traits, with highlighted distinctions between ascending aorta and aortic arch. The lower part of the figure recapitulates the detailed aortic root components: the sinotubular junction of aortic root with ascending aorta; the aortic valve leaflets; the ventriculo‐aortic junction of aortic root and left heart ventricle.
Figure 2. Figure 2. Echocardiography of normal and stenotic aortic valve in systole. 2D‐echocardiography of a healthy (left) and a stenotic (right) aortic valve in short‐axis view. Red arrows (left) highlight the normal, thin leaflets with an optimal orifice area. Red stars (right) highlight thick calcific leaflet edges with a restricted aortic valve area.
Figure 3. Figure 3. Cellular features of stenotic aortic valve. The cartoon depicts the main cellular mechanisms involved in the pathogenesis of aortic valve stenosis. (A) The differentiation of monocytes into osteoclasts. (B) The endothelial damage leading to endothelial‐to‐mesenchymal transition (EndMT) into activated myofibroblasts. (C) The differentiation of valve interstitial cells (VIC) into activated myofibroblast and/or osteoblast‐like cells, leading to fibrosis and calcification, respectively. Adapted with permission from (132).
Figure 4. Figure 4. Aortic valve stenosis pathophysiology. Phenotype of stenotic aortic valves and the involvement of valve stenosis in cardiac diseases. In the left side are depicted normal and stenotic aortic valves in diastole and systole. In the stenotic picture are present several leaflet regions of spotty calcification, which stiffen the valve structure and alter the dynamic of valve opening. In the right side of the figure are mentioned the main cardiac pathologies caused by aortic valve stenosis.
Figure 5. Figure 5. Mitral valve anatomy. Structure and nomenclature of mitral valve. The mitral valve represents the gate between left atrium and left ventricle. The perfect closure of the valve depends on cardiac papillary muscles, which orchestrate the leaflet traction during systole through chordae tendinae.
Figure 6. Figure 6. Echocardiography of healthy and prolapsed mitral valve in systole. 2D‐echocardiography of a healthy (left) and regurgitant (central and right) mitral valve in long axis view. The left echocardiography image shows the correct alignment (coaptation—red arrow) of healthy, thin mitral valve leaflets. The central echocardiography shows impaired closure (red star). The right color‐Doppler image shows the regurgitant volume toward the left atrium.
Figure 7. Figure 7. Cellular features of mitral valve regurgitation. The cartoon depicts the main cellular mechanisms involved in the pathogenesis of mitral valve regurgitation. (A) The endothelial damage leading to EndMT of VECs into myofibroblas (MyoFB). (B) The fragmentation of elastin in the atrialis layer. (C) The differentiation of VICs into MyoFB leading to fibrosis (collagen and glycosaminoglycans deposition depicted in blue in the spongiosa layer). (D) The collagen fragmentation in the fibrosa layer. Adapted with permission from (109).
Figure 8. Figure 8. Mitral valve regurgitation pathophysiology. Phenotype of regurgitant mitral valve and its involvement in cardiac diseases. In the left side pictures of normal and regurgitant mitral valve in systole and diastole. In the closed regurgitant mitral valve picture is clear the improper coaptation of valve leaflets. In the right side of the figure are mentioned the main cardiac pathologies caused by mitral valve regurgitation.


Figure 1. Aortic valve anatomy. Structure and nomenclature of aortic root components. The upper side of the figure represents thoracic aortic traits, with highlighted distinctions between ascending aorta and aortic arch. The lower part of the figure recapitulates the detailed aortic root components: the sinotubular junction of aortic root with ascending aorta; the aortic valve leaflets; the ventriculo‐aortic junction of aortic root and left heart ventricle.


Figure 2. Echocardiography of normal and stenotic aortic valve in systole. 2D‐echocardiography of a healthy (left) and a stenotic (right) aortic valve in short‐axis view. Red arrows (left) highlight the normal, thin leaflets with an optimal orifice area. Red stars (right) highlight thick calcific leaflet edges with a restricted aortic valve area.


Figure 3. Cellular features of stenotic aortic valve. The cartoon depicts the main cellular mechanisms involved in the pathogenesis of aortic valve stenosis. (A) The differentiation of monocytes into osteoclasts. (B) The endothelial damage leading to endothelial‐to‐mesenchymal transition (EndMT) into activated myofibroblasts. (C) The differentiation of valve interstitial cells (VIC) into activated myofibroblast and/or osteoblast‐like cells, leading to fibrosis and calcification, respectively. Adapted with permission from (132).


Figure 4. Aortic valve stenosis pathophysiology. Phenotype of stenotic aortic valves and the involvement of valve stenosis in cardiac diseases. In the left side are depicted normal and stenotic aortic valves in diastole and systole. In the stenotic picture are present several leaflet regions of spotty calcification, which stiffen the valve structure and alter the dynamic of valve opening. In the right side of the figure are mentioned the main cardiac pathologies caused by aortic valve stenosis.


Figure 5. Mitral valve anatomy. Structure and nomenclature of mitral valve. The mitral valve represents the gate between left atrium and left ventricle. The perfect closure of the valve depends on cardiac papillary muscles, which orchestrate the leaflet traction during systole through chordae tendinae.


Figure 6. Echocardiography of healthy and prolapsed mitral valve in systole. 2D‐echocardiography of a healthy (left) and regurgitant (central and right) mitral valve in long axis view. The left echocardiography image shows the correct alignment (coaptation—red arrow) of healthy, thin mitral valve leaflets. The central echocardiography shows impaired closure (red star). The right color‐Doppler image shows the regurgitant volume toward the left atrium.


Figure 7. Cellular features of mitral valve regurgitation. The cartoon depicts the main cellular mechanisms involved in the pathogenesis of mitral valve regurgitation. (A) The endothelial damage leading to EndMT of VECs into myofibroblas (MyoFB). (B) The fragmentation of elastin in the atrialis layer. (C) The differentiation of VICs into MyoFB leading to fibrosis (collagen and glycosaminoglycans deposition depicted in blue in the spongiosa layer). (D) The collagen fragmentation in the fibrosa layer. Adapted with permission from (109).


Figure 8. Mitral valve regurgitation pathophysiology. Phenotype of regurgitant mitral valve and its involvement in cardiac diseases. In the left side pictures of normal and regurgitant mitral valve in systole and diastole. In the closed regurgitant mitral valve picture is clear the improper coaptation of valve leaflets. In the right side of the figure are mentioned the main cardiac pathologies caused by mitral valve regurgitation.
References
 1.Acker MA. Should moderate or greater mitral regurgitation be repaired in all patients with LVEF <30%? Mitral valve repair in patients with advanced heart failure and severe functional mitral insufficiency reverses left ventricular remodeling and improves symptoms. Circ Heart Fail 1: 281‐284, 2008.
 2.Adams DH, Rosenhek R, Falk V. Degenerative mitral valve regurgitation: Best practice revolution. Eur Heart J 31: 1958‐1966, 2010.
 3.Aikawa E, Nahrendorf M, Figueiredo JL, Swirski FK, Shtatland T, Kohler RH, Jaffer FA, Aikawa M, Weissleder R. Osteogenesis associates with inflammation in early‐stage atherosclerosis evaluated by molecular imaging in vivo. Circulation 116: 2841‐2850, 2007.
 4.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 115: 377‐386, 2007.
 5.Al‐Aly Z, Shao JS, Lai CF, Huang E, Cai J, Behrmann A, Cheng SL, Towler DA. Aortic Msx2‐Wnt calcification cascade is regulated by TNF‐alpha‐dependent signals in diabetic Ldlr−/− mice. Arterioscler Thromb Vasc Biol 27: 2589‐2596, 2007.
 6.Alexopoulos A, Bravou V, Peroukides S, Kaklamanis L, Varakis J, Alexopoulos D, Papadaki H. Bone regulatory factors NFATc1 and Osterix in human calcific aortic valves. Int J Cardiol 139: 142‐149, 2010.
 7.Ankeny RF, Thourani VH, Weiss D, Vega JD, Taylor WR, Nerem RM, Jo H. Preferential activation of SMAD1/5/8 on the fibrosa endothelium in calcified human aortic valves‐–association with low BMP antagonists and SMAD6. PLoS One 6: e20969, 2011.
 8.Ann EJ, Kim HY, Choi YH, Kim MY, Mo JS, Jung J, Yoon JH, Kim SM, Moon JS, Seo MS, Hong JA, Jang WG, Shore P, Komori T, Koh JT, Park HS. Inhibition of Notch1 signaling by Runx2 during osteoblast differentiation. J Bone Miner Res 26: 317‐330, 2011.
 9.Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta activation. J Cell Sci 116: 217‐224, 2003.
 10.Arishiro K, Hoshiga M, Negoro N, Jin D, Takai S, Miyazaki M, Ishihara T, Hanafusa T. Angiotensin receptor‐1 blocker inhibits atherosclerotic changes and endothelial disruption of the aortic valve in hypercholesterolemic rabbits. J Am Coll Cardiol 49: 1482‐1489, 2007.
 11.Askevold ET, Gullestad L, Aakhus S, Ranheim T, Tonnessen T, Solberg OG, Aukrust P, Ueland T. Secreted Wnt modulators in symptomatic aortic stenosis. J Am Heart Assoc 1: e002261, 2012.
 12.Aupperle H, Marz I, Thielebein J, Schoon HA. Expression of transforming growth factor‐beta1, ‐beta2 and ‐beta3 in normal and diseased canine mitral valves. J Comp Pathol 139: 97‐107, 2008.
 13.Barber JE, Kasper FK, Ratliff NB, Cosgrove DM, Griffin BP, Vesely I. Mechanical properties of myxomatous mitral valves. J Thorac Cardiovasc Surg 122: 955‐962, 2001.
 14.Basta G, Corciu AI, Vianello A, Del Turco S, Foffa I, Navarra T, Chiappino D, Berti S, Mazzone A. Circulating soluble receptor for advanced glycation end‐product levels are decreased in patients with calcific aortic valve stenosis. Atherosclerosis 210: 614‐618, 2010.
 15.Baumgartner H, Hung J, Bermejo J, Chambers JB, Evangelista A, Griffin BP, Iung B, Otto CM, Pellikka PA, Quinones M. Echocardiographic assessment of valve stenosis: EAE/ASE recommendations for clinical practice. Eur J Echocardiogr 10: 1‐25, 2009.
 16.Bennett BJ, Scatena M, Kirk EA, Rattazzi M, Varon RM, Averill M, Schwartz SM, Giachelli CM, Rosenfeld ME. Osteoprotegerin inactivation accelerates advanced atherosclerotic lesion progression and calcification in older ApoE−/− mice. Arterioscler Thromb Vasc Biol 26: 2117‐2124, 2006.
 17.Bonow RO, Carabello BA, Kanu C, de Leon AC, Jr., Faxon DP, Freed MD, Gaasch WH, Lytle BW, Nishimura RA, O'Gara PT, O'Rourke RA, Otto CM, Shah PM, Shanewise JS, Smith SC, Jr., Jacobs AK, Adams CD, Anderson JL, Antman EM, Fuster V, Halperin JL, Hiratzka LF, Hunt SA, Nishimura R, Page RL, Riegel B. ACC/AHA 2006 guidelines for the management of patients with valvular heart disease: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (writing committee to revise the 1998 Guidelines for the Management of Patients With Valvular Heart Disease): Developed in collaboration with the Society of Cardiovascular Anesthesiologists: endorsed by the Society for Cardiovascular Angiography and Interventions and the Society of Thoracic Surgeons. Circulation 114: e84‐231, 2006.
 18.Bostrom KI, Rajamannan NM, Towler DA. The regulation of valvular and vascular sclerosis by osteogenic morphogens. Circ Res 109: 564‐577, 2011.
 19.Boudoulas H. Etiology of valvular heart disease. Expert Rev Cardiovasc Ther 1: 523‐532, 2003.
 20.Breithardt OA, Sinha AM, Schwammenthal E, Bidaoui N, Markus KU, Franke A, Stellbrink C. Acute effects of cardiac resynchronization therapy on functional mitral regurgitation in advanced systolic heart failure. J Am Coll Cardiol 41: 765‐770, 2003.
 21.Bursi F, Enriquez‐Sarano M, Jacobsen SJ, Roger VL. Mitral regurgitation after myocardial infarction: a review. Am J Med 119: 103‐112, 2006.
 22.Butcher JT, Nerem RM. Valvular endothelial cells regulate the phenotype of interstitial cells in co‐culture: Effects of steady shear stress. Tissue Eng 12: 905‐915, 2006.
 23.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 24: 1429‐1434, 2004.
 24.Butcher JT, Simmons CA, Warnock JN. Mechanobiology of the aortic heart valve. J Heart Valve Dis 17: 62‐73, 2008.
 25.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 26: 69‐77, 2006.
 26.Byon CH, Sun Y, Chen J, Yuan K, Mao X, Heath JM, Anderson PG, Tintut Y, Demer LL, Wang D, Chen Y. Runx2‐upregulated receptor activator of nuclear factor kappaB ligand in calcifying smooth muscle cells promotes migration and osteoclastic differentiation of macrophages. Arterioscler Thromb Vasc Biol 31: 1387‐1396, 2011.
 27.Cagirci G, Cay S, Canga A, Karakurt O, Yazihan N, Kilic H, Topaloglu S, Aras D, Demir AD, Akdemir R. Association between plasma asymmetrical dimethylarginine activity and severity of aortic valve stenosis. J Cardiovasc Med (Hagerstown) 12: 96‐101, 2011.
 28.Caira FC, Stock SR, Gleason TG, McGee EC, Huang J, Bonow RO, Spelsberg TC, McCarthy PM, Rahimtoola SH, Rajamannan NM. Human degenerative valve disease is associated with up‐regulation of low‐density lipoprotein receptor‐related protein 5 receptor‐mediated bone formation. J Am Coll Cardiol 47: 1707‐1712, 2006.
 29.Capomolla S, Febo O, Gnemmi M, Riccardi G, Opasich C, Caporotondi A, Mortara A, Pinna GD, Cobelli F. Beta‐blockade therapy in chronic heart failure: Diastolic function and mitral regurgitation improvement by carvedilol. Am Heart J 139: 596‐608, 2000.
 30.Carabello BA, Paulus WJ. Aortic stenosis. Lancet 373: 956‐966, 2009.
 31.Carpentier A. Cardiac valve surgery–‐the “French correction”. J Thorac Cardiovasc Surg 86: 323‐337, 1983.
 32.Carpentier AF, Lessana A, Relland JY, Belli E, Mihaileanu S, Berrebi AJ, Palsky E, Loulmet DF. The “physio‐ring”: An advanced concept in mitral valve annuloplasty. Ann Thorac Surg 60: 1177‐1185; discussion 1185‐1176, 1995.
 33.Cary T, Pearce J. Aortic stenosis: Pathophysiology, diagnosis, and medical management of nonsurgical patients. Crit Care Nurse 33: 58‐72, 2013.
 34.Chakraborty S, Wirrig EE, Hinton RB, Merrill WH, Spicer DB, Yutzey KE. Twist1 promotes heart valve cell proliferation and extracellular matrix gene expression during development in vivo and is expressed in human diseased aortic valves. Dev Biol 347: 167‐179, 2010.
 35.Chang K, Weiss D, Suo J, Vega JD, Giddens D, Taylor WR, Jo H. Bone morphogenic protein antagonists are coexpressed with bone morphogenic protein 4 in endothelial cells exposed to unstable flow in vitro in mouse aortas and in human coronary arteries: Role of bone morphogenic protein antagonists in inflammation and atherosclerosis. Circulation 116: 1258‐1266, 2007.
 36.Chen D, Zhao M, Mundy GR. Bone morphogenetic proteins. Growth Factors 22: 233‐241, 2004.
 37.Chen JH, Simmons CA. Cell‐matrix interactions in the pathobiology of calcific aortic valve disease: Critical roles for matricellular, matricrine, and matrix mechanics cues. Circ Res 108: 1510‐1524, 2011.
 38.Combs MD, Yutzey KE. Heart valve development: Regulatory networks in development and disease. Circ Res 105: 408‐421, 2009.
 39.Corcoran BM, Black A, Anderson H, McEwan JD, French A, Smith P, Devine C. Identification of surface morphologic changes in the mitral valve leaflets and chordae tendineae of dogs with myxomatous degeneration. Am J Vet Res 65: 198‐206, 2004.
 40.Cowell SJ, Newby DE, Prescott RJ, Bloomfield P, Reid J, Northridge DB, Boon NA. A randomized trial of intensive lipid‐lowering therapy in calcific aortic stenosis. N Engl J Med 352: 2389‐2397, 2005.
 41.Cucoranu I, Clempus R, Dikalova A, Phelan PJ, Ariyan S, Dikalov S, Sorescu D. NAD(P)H oxidase 4 mediates transforming growth factor‐beta1‐induced differentiation of cardiac fibroblasts into myofibroblasts. Circ Res 97: 900‐907, 2005.
 42.Cushing MC, Liao JT, Anseth KS. Activation of valvular interstitial cells is mediated by transforming growth factor‐beta1 interactions with matrix molecules. Matrix Biol 24: 428‐437, 2005.
 43.D'Amelio P, Isaia G, Isaia GC. The osteoprotegerin/RANK/RANKL system: A bone key to vascular disease. J Endocrinol Invest 32: 6‐9, 2009.
 44.Dal‐Bianco JP, Levine RA. Anatomy of the mitral valve apparatus: Role of 2D and 3D echocardiography. Cardiol Clin 31: 151‐164, 2013.
 45.Delling FN, Vasan RS. Epidemiology and pathophysiology of mitral valve prolapse: New insights into disease progression, genetics, and molecular basis. Circulation 129: 2158‐2170, 2014.
 46.Disatian S, Ehrhart EJ, III, Zimmerman S, Orton EC. Interstitial cells from dogs with naturally occurring myxomatous mitral valve disease undergo phenotype transformation. J Heart Valve Dis 17: 402‐411; discussion 412, 2008.
 47.Disatian S, Lacerda C, Orton EC. Tryptophan hydroxylase 1 expression is increased in phenotype‐altered canine and human degenerative myxomatous mitral valves. J Heart Valve Dis 19: 71‐78, 2010.
 48.Disatian S, Orton EC. Autocrine serotonin and transforming growth factor beta 1 signaling mediates spontaneous myxomatous mitral valve disease. J Heart Valve Dis 18: 44‐51, 2009.
 49.Droogmans S, Roosens B, Cosyns B, Degaillier C, Hernot S, Weytjens C, Garbar C, Caveliers V, Pipeleers‐Marichal M, Franken PR, Bossuyt A, Schoors D, Lahoutte T, Van Camp G. Dose dependency and reversibility of serotonin‐induced valvular heart disease in rats. Cardiovasc Toxicol 9: 134‐141, 2009.
 50.Edep ME, Shirani J, Wolf P, Brown DL. Matrix metalloproteinase expression in nonrheumatic aortic stenosis. Cardiovasc Pathol 9: 281‐286, 2000.
 51.Elangbam CS, Job LE, Zadrozny LM, Barton JC, Yoon LW, Gates LD, Slocum N. 5‐hydroxytryptamine (5HT)‐induced valvulopathy: Compositional valvular alterations are associated with 5HT2B receptor and 5HT transporter transcript changes in Sprague‐Dawley rats. Exp Toxicol Pathol 60: 253‐262, 2008.
 52.Enriquez‐Sarano M, Akins CW, Vahanian A. Mitral regurgitation. Lancet 373: 1382‐1394, 2009.
 53.Fischer L, Boland G, Tuan RS. Wnt‐3A enhances bone morphogenetic protein‐2‐mediated chondrogenesis of murine C3H10T1/2 mesenchymal cells. J Biol Chem 277: 30870‐30878, 2002.
 54.Fitzgerald LW, Burn TC, Brown BS, Patterson JP, Corjay MH, Valentine PA, Sun JH, Link JR, Abbaszade I, Hollis JM, Largent BL, Hartig PR, Hollis GF, Meunier PC, Robichaud AJ, Robertson DW. Possible role of valvular serotonin 5‐HT(2B) receptors in the cardiopathy associated with fenfluramine. Mol Pharmacol 57: 75‐81, 2000.
 55.Fondard O, Detaint D, Iung B, Choqueux C, Adle‐Biassette H, Jarraya M, Hvass U, Couetil JP, Henin D, Michel JB, Vahanian A, Jacob MP. Extracellular matrix remodelling in human aortic valve disease: The role of matrix metalloproteinases and their tissue inhibitors. Eur Heart J 26: 1333‐1341, 2005.
 56.Freeman RV, Otto CM. Spectrum of calcific aortic valve disease: Pathogenesis, disease progression, and treatment strategies. Circulation 111: 3316‐3326, 2005.
 57.Gaasch WH, Meyer TE. Left ventricular response to mitral regurgitation: Implications for management. Circulation 118: 2298‐2303, 2008.
 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 24: 171‐174, 2000.
 59.Garg V, Muth AN, Ransom JF, Schluterman MK, Barnes R, King IN, Grossfeld PD, Srivastava D. Mutations in NOTCH1 cause aortic valve disease. Nature 437: 270‐274, 2005.
 60.Gawdzik J, Mathew L, Kim G, Puri TS, Hofmann Bowman MA. Vascular remodeling and arterial calcification are directly mediated by S100A12 (EN‐RAGE) in chronic kidney disease. Am J Nephrol 33: 250‐259, 2011.
 61.Geirsson A, Singh M, Ali R, Abbas H, Li W, Sanchez JA, Hashim S, Tellides G. Modulation of transforming growth factor‐beta signaling and extracellular matrix production in myxomatous mitral valves by angiotensin II receptor blockers. Circulation 126: S189‐197, 2012.
 62.Geroldi D, Falcone C, Emanuele E. Soluble receptor for advanced glycation end products: From disease marker to potential therapeutic target. Curr Med Chem 13: 1971‐1978, 2006.
 63.Gould KL, Carabello BA. Why angina in aortic stenosis with normal coronary arteriograms? Circulation 107: 3121‐3123, 2003.
 64.Grewal JS, Mukhin YV, Garnovskaya MN, Raymond JR, Greene EL. Serotonin 5‐HT2A receptor induces TGF‐beta1 expression in mesangial cells via ERK: Proliferative and fibrotic signals. Am J Physiol 276: F922‐F930, 1999.
 65.Guerraty MA, Grant GR, Karanian JW, Chiesa OA, Pritchard WF, Davies PF. Hypercholesterolemia induces side‐specific phenotypic changes and peroxisome proliferator‐activated receptor‐gamma pathway activation in swine aortic valve endothelium. Arterioscler Thromb Vasc Biol 30: 225‐231, 2010.
 66.Gupta V, Werdenberg JA, Lawrence BD, Mendez JS, Stephens EH, Grande‐Allen KJ. Reversible secretion of glycosaminoglycans and proteoglycans by cyclically stretched valvular cells in 3D culture. Ann Biomed Eng 36: 1092‐1103, 2008.
 67.Gustafsson BI, Hauso O, Drozdov I, Kidd M, Modlin IM. Carcinoid heart disease. Int J Cardiol 129: 318‐324, 2008.
 68.Gustafsson BI, Tommeras K, Nordrum I, Loennechen JP, Brunsvik A, Solligard E, Fossmark R, Bakke I, Syversen U, Waldum H. Long‐term serotonin administration induces heart valve disease in rats. Circulation 111: 1517‐1522, 2005.
 69.Hafizi S, Taylor PM, Chester AH, Allen SP, Yacoub MH. Mitogenic and secretory responses of human valve interstitial cells to vasoactive agents. J Heart Valve Dis 9: 454‐458, 2000.
 70.Hagler MA, Hadley TM, Zhang H, Mehra K, Roos CM, Schaff HV, Suri RM, Miller JD. TGF‐beta signalling and reactive oxygen species drive fibrosis and matrix remodelling in myxomatous mitral valves. Cardiovasc Res 99: 175‐184, 2013.
 71.Hamblin M, Chang L, Fan Y, Zhang J, Chen YE. PPARs and the cardiovascular system. Antioxid Redox Signal 11: 1415‐1452, 2009.
 72.Hartmann C, Tabin CJ. Dual roles of Wnt signaling during chondrogenesis in the chicken limb. Development 127: 3141‐3159, 2000.
 73.Hein S, Arnon E, Kostin S, Schonburg M, Elsasser A, Polyakova V, Bauer EP, Klovekorn WP, Schaper J. Progression from compensated hypertrophy to failure in the pressure‐overloaded human heart: Structural deterioration and compensatory mechanisms. Circulation 107: 984‐991, 2003.
 74.Heistad DD, Wakisaka Y, Miller J, Chu Y, Pena‐Silva R. Novel aspects of oxidative stress in cardiovascular diseases. Circ J 73: 201‐207, 2009.
 75.Helske S, Lindstedt KA, Laine M, Mayranpaa M, Werkkala K, Lommi J, Turto H, Kupari M, Kovanen PT. Induction of local angiotensin II‐producing systems in stenotic aortic valves. J Am Coll Cardiol 44: 1859‐1866, 2004.
 76.Helske S, Syvaranta S, Kupari M, Lappalainen J, Laine M, Lommi J, Turto H, Mayranpaa M, Werkkala K, Kovanen PT, Lindstedt KA. Possible role for mast cell‐derived cathepsin G in the adverse remodelling of stenotic aortic valves. Eur Heart J 27: 1495‐1504, 2006.
 77.Helske S, Syvaranta S, Lindstedt KA, Lappalainen J, Oorni K, Mayranpaa MI, Lommi J, Turto H, Werkkala K, Kupari M, Kovanen PT. Increased expression of elastolytic cathepsins S, K, and V and their inhibitor cystatin C in stenotic aortic valves. Arterioscler Thromb Vasc Biol 26: 1791‐1798, 2006.
 78.Hess OM, Villari B, Krayenbuehl HP. Diastolic dysfunction in aortic stenosis. Circulation 87: IV73‐IV76, 1993.
 79.Hofmann Bowman MA, Gawdzik J, Bukhari U, Husain AN, Toth PT, Kim G, Earley J, McNally EM. S100A12 in vascular smooth muscle accelerates vascular calcification in apolipoprotein E‐null mice by activating an osteogenic gene regulatory program. Arterioscler Thromb Vasc Biol 31: 337‐344, 2011.
 80.Hofmann C, Penner U, Dorow R, Pertz HH, Jahnichen S, Horowski R, Latte KP, Palla D, Schurad B. Lisuride, a dopamine receptor agonist with 5‐HT2B receptor antagonist properties: absence of cardiac valvulopathy adverse drug reaction reports supports the concept of a crucial role for 5‐HT2B receptor agonism in cardiac valvular fibrosis. Clin Neuropharmacol 29: 80‐86, 2006.
 81.Huang F, Chen YG. Regulation of TGF‐beta receptor activity. Cell Biosci 2: 9, 2012.
 82.Hulin A, Deroanne CF, Lambert CA, Dumont B, Castronovo V, Defraigne JO, Nusgens BV, Radermecker MA, Colige AC. Metallothionein‐dependent up‐regulation of TGF‐beta2 participates in the remodelling of the myxomatous mitral valve. Cardiovasc Res 93: 480‐489, 2012.
 83.Hurlstone AF, Haramis AP, Wienholds E, Begthel H, Korving J, Van Eeden F, Cuppen E, Zivkovic D, Plasterk RH, Clevers H. The Wnt/beta‐catenin pathway regulates cardiac valve formation. Nature 425: 633‐637, 2003.
 84.Hyytiainen M, Penttinen C, Keski‐Oja J. Latent TGF‐beta binding proteins: Extracellular matrix association and roles in TGF‐beta activation. Crit Rev Clin Lab Sci 41: 233‐264, 2004.
 85.Isoda K, Matsuki T, Kondo H, Iwakura Y, Ohsuzu F. Deficiency of interleukin‐1 receptor antagonist induces aortic valve disease in BALB/c mice. Arterioscler Thromb Vasc Biol 30: 708‐715, 2010.
 86.Iung B, Baron G, Butchart EG, Delahaye F, Gohlke‐Barwolf C, Levang OW, Tornos P, Vanoverschelde JL, Vermeer F, Boersma E, Ravaud P, Vahanian A. A prospective survey of patients with valvular heart disease in Europe: The Euro Heart Survey on Valvular Heart Disease. Eur Heart J 24: 1231‐1243, 2003.
 87.Jameson AG. Pathologic physiology in mitral valve disease. Ann N Y Acad Sci 118: 486‐489, 1965.
 88.Jian B, Jones PL, Li Q, Mohler ER, III, Schoen FJ, Levy RJ. Matrix metalloproteinase‐2 is associated with tenascin‐C in calcific aortic stenosis. Am J Pathol 159: 321‐327, 2001.
 89.Jian B, Narula N, Li QY, Mohler ER, III, 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 75: 457‐465; discussion 465‐456, 2003.
 90.Jian B, Xu J, Connolly J, Savani RC, Narula N, Liang B, Levy RJ. Serotonin mechanisms in heart valve disease I: Serotonin‐induced up‐regulation of transforming growth factor‐beta1 via G‐protein signal transduction in aortic valve interstitial cells. Am J Pathol 161: 2111‐2121, 2002.
 91.Johnson RC, Leopold JA, Loscalzo J. Vascular calcification: Pathobiological mechanisms and clinical implications. Circ Res 99: 1044‐1059, 2006.
 92.Kaden JJ, Bickelhaupt S, Grobholz R, Haase KK, Sarikoc A, Kilic R, Brueckmann M, Lang S, Zahn I, Vahl C, Hagl S, Dempfle CE, Borggrefe M. Receptor activator of nuclear factor kappaB ligand and osteoprotegerin regulate aortic valve calcification. J Mol Cell Cardiol 36: 57‐66, 2004.
 93.Kaden JJ, Dempfle CE, Grobholz R, Fischer CS, Vocke DC, Kilic R, Sarikoc A, Pinol R, Hagl S, Lang S, Brueckmann M, Borggrefe M. Inflammatory regulation of extracellular matrix remodeling in calcific aortic valve stenosis. Cardiovasc Pathol 14: 80‐87, 2005.
 94.Kamath AR, Pai RG. Risk factors for progression of calcific aortic stenosis and potential therapeutic targets. Int J Angiol 17: 63‐70, 2008.
 95.Kapoor WN. Syncope. In: Walker HK, Hall WD, Hurst JW, editors. Clinical Methods: The History, Physical, and Laboratory Examinations. Boston: 1990.
 96.Katz R, Budoff MJ, Takasu J, Shavelle DM, Bertoni A, Blumenthal RS, Ouyang P, Wong ND, O'Brien KD. Relationship of metabolic syndrome with incident aortic valve calcium and aortic valve calcium progression: The Multi‐Ethnic Study of Atherosclerosis (MESA). Diabetes 58: 813‐819, 2009.
 97.Kawaguchi H, Akune T, Yamaguchi M, Ohba S, Ogata N, Chung UI, Kubota N, Terauchi Y, Kadowaki T, Nakamura K. Distinct effects of PPARgamma insufficiency on bone marrow cells, osteoblasts, and osteoclastic cells. J Bone Miner Metab 23: 275‐279, 2005.
 98.Keen HL, Ryan MJ, Beyer A, Mathur S, Scheetz TE, Gackle BD, Faraci FM, Casavant TL, Sigmund CD. Gene expression profiling of potential PPARgamma target genes in mouse aorta. Physiol Genomics 18: 33‐42, 2004.
 99.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 602: 28‐35, 2009.
 100.Khan R, Sheppard R. Fibrosis in heart disease: Understanding the role of transforming growth factor‐beta in cardiomyopathy, valvular disease and arrhythmia. Immunology 118: 10‐24, 2006.
 101.Kiefer TL, Bashore TM. Pulmonary hypertension related to left‐sided cardiac pathology. Pulm Med 2011: 381787, 2011.
 102.Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci 71: 549‐574, 2014.
 103.Kopuz C, Erk K, Baris YS, Onderoglu S, Sinav A. Morphometry of the fibrous ring of the mitral valve. Ann Anat 177: 151‐154, 1995.
 104.Kupfahl C, Pink D, Friedrich K, Zurbrugg HR, Neuss M, Warnecke C, Fielitz J, Graf K, Fleck E, Regitz‐Zagrosek V. Angiotensin II directly increases transforming growth factor beta1 and osteopontin and indirectly affects collagen mRNA expression in the human heart. Cardiovasc Res 46: 463‐475, 2000.
 105.Kurtz CE, Otto CM. Aortic stenosis: Clinical aspects of diagnosis and management, with 10 illustrative case reports from a 25‐year experience. Medicine (Baltimore) 89: 349‐379, 2010.
 106.Lacerda CM, Maclea HB, Kisiday JD, Orton EC. Static and cyclic tensile strain induce myxomatous effector proteins and serotonin in canine mitral valves. J Vet Cardiol 14: 223‐230, 2012.
 107.Launay JM, Birraux G, Bondoux D, Callebert J, Choi DS, Loric S, Maroteaux L. Ras involvement in signal transduction by the serotonin 5‐HT2B receptor. J Biol Chem 271: 3141‐3147, 1996.
 108.Lee NK, Choi YG, Baik JY, Han SY, Jeong DW, Bae YS, Kim N, Lee SY. A crucial role for reactive oxygen species in RANKL‐induced osteoclast differentiation. Blood 106: 852‐859, 2005.
 109.Levine RA, Hagege AA, Judge DP, Padala M, Dal‐Bianco JP, Aikawa E, Beaudoin J, Bischoff J, Bouatia‐Naji N, Bruneval P, Butcher JT, Carpentier A, Chaput M, Chester AH, Clusel C, Delling FN, Dietz HC, Dina C, Durst R, Fernandez‐Friera L, Handschumacher MD, Jensen MO, Jeunemaitre XP, Le Marec H, Le Tourneau T, Markwald RR, Merot J, Messas E, Milan DP, Neri T, Norris RA, Peal D, Perrocheau M, Probst V, Puceat M, Rosenthal N, Solis J, Schott JJ, Schwammenthal E, Slaugenhaupt SA, Song JK, Yacoub MH. Mitral valve disease‐‐‐morphology and mechanisms. Nat Rev Cardiol 12: 689‐710, 2015.
 110.Levine RA, Handschumacher MD, Sanfilippo AJ, Hagege AA, Harrigan P, Marshall JE, Weyman AE. Three‐dimensional echocardiographic reconstruction of the mitral valve, with implications for the diagnosis of mitral valve prolapse. Circulation 80: 589‐598, 1989.
 111.Levine RA, Triulzi MO, Harrigan P, Weyman AE. The relationship of mitral annular shape to the diagnosis of mitral valve prolapse. Circulation 75: 756‐767, 1987.
 112.Lincoln J, Lange AW, Yutzey KE. Hearts and bones: Shared regulatory mechanisms in heart valve, cartilage, tendon, and bone development. Dev Biol 294: 292‐302, 2006.
 113.Linde C, Leclercq C, Rex S, Garrigue S, Lavergne T, Cazeau S, McKenna W, Fitzgerald M, Deharo JC, Alonso C, Walker S, Braunschweig F, Bailleul C, Daubert JC. Long‐term benefits of biventricular pacing in congestive heart failure: Results from the MUltisite STimulation in cardiomyopathy (MUSTIC) study. J Am Coll Cardiol 40: 111‐118, 2002.
 114.Liu AC, Joag VR, Gotlieb AI. The emerging role of valve interstitial cell phenotypes in regulating heart valve pathobiology. Am J Pathol 171: 1407‐1418, 2007.
 115.Liu Y, Suzuki YJ, Day RM, Fanburg BL. Rho kinase‐induced nuclear translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused by serotonin. Circ Res 95: 579‐586, 2004.
 116.Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol 20: 781‐810, 2004.
 117.Luna‐Zurita L, Prados B, Grego‐Bessa J, Luxan G, del Monte G, Benguria A, Adams RH, Perez‐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 120: 3493‐3507, 2010.
 118.Luo S, Lei H, Qin H, Xia Y. Molecular mechanisms of endothelial NO synthase uncoupling. Curr Pharm Des 20: 3548‐3553, 2014.
 119.Lyons RM, Keski‐Oja J, Moses HL. Proteolytic activation of latent transforming growth factor‐beta from fibroblast‐conditioned medium. J Cell Biol 106: 1659‐1665, 1988.
 120.MacDonald BT, Tamai K, He X. Wnt/beta‐catenin signaling: Components, mechanisms, and diseases. Dev Cell 17: 9‐26, 2009.
 121.MacGrogan D, Luna‐Zurita L, de la Pompa JL. Notch signaling in cardiac valve development and disease. Birth Defects Res Part A Clin Mol Teratol 91: 449‐459, 2011.
 122.MacLea HB, Boon JA, Bright JM. Doppler echocardiographic evaluation of midventricular obstruction in cats with hypertrophic cardiomyopathy. J Vet Intern Med 27: 1416‐1420, 2013.
 123.Macsai CE, Foster BK, Xian CJ. Roles of Wnt signalling in bone growth, remodelling, skeletal disorders and fracture repair. J Cell Physiol 215: 578‐587, 2008.
 124.Madu EC, D'Cruz IA. The vital role of papillary muscles in mitral and ventricular function: Echocardiographic insights. Clin Cardiol 20: 93‐98, 1997.
 125.Marchesi C, Paradis P, Schiffrin EL. Role of the renin‐angiotensin system in vascular inflammation. Trends Pharmacol Sci 29: 367‐374, 2008.
 126.Markwald RR, Butcher JT. The next frontier in cardiovascular developmental biology–‐an integrated approach to adult disease? Nat Clin Pract Cardiovasc Med 4: 60‐61, 2007.
 127.Markwald RR, Norris RA, Moreno‐Rodriguez R, Levine RA. Developmental basis of adult cardiovascular diseases: Valvular heart diseases. Ann N Y Acad Sci 1188: 177‐183, 2010.
 128.McCarthy KP, Ring L, Rana BS. Anatomy of the mitral valve: Understanding the mitral valve complex in mitral regurgitation. Eur J Echocardiogr 11: i3‐i9, 2010.
 129.Mekontso‐Dessap A, Brouri F, Pascal O, Lechat P, Hanoun N, Lanfumey L, Seif I, Benhaiem‐Sigaux N, Kirsch M, Hamon M, Adnot S, Eddahibi S. Deficiency of the 5‐hydroxytryptamine transporter gene leads to cardiac fibrosis and valvulopathy in mice. Circulation 113: 81‐89, 2006.
 130.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 16: 268‐276, 2007.
 131.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 52: 843‐850, 2008.
 132.Miller JD, Weiss RM, Heistad DD. Calcific aortic valve stenosis: Methods, models, and mechanisms. Circ Res 108: 1392‐1412, 2011.
 133.Miller JD, Weiss RM, Serrano KM, Brooks RM, II, Berry CJ, Zimmerman K, Young SG, Heistad DD. Lowering plasma cholesterol levels halts progression of aortic valve disease in mice. Circulation 119: 2693‐2701, 2009.
 134.Miller JD, Weiss RM, Serrano KM, Castaneda LE, Brooks RM, Zimmerman K, Heistad DD. Evidence for active regulation of pro‐osteogenic signaling in advanced aortic valve disease. Arterioscler Thromb Vasc Biol 30: 2482‐2486, 2010.
 135.Miyazono K, Hellman U, Wernstedt C, Heldin CH. Latent high molecular weight complex of transforming growth factor beta 1. Purification from human platelets and structural characterization. J Biol Chem 263: 6407‐6415, 1988.
 136.Moesgaard SG, Olsen LH, Aasted B, Viuff BM, Pedersen LG, Pedersen HD, Harrison AP. Direct measurements of nitric oxide release in relation to expression of endothelial nitric oxide synthase in isolated porcine mitral valves. J Vet Med A Physiol Pathol Clin Med 54: 156‐160, 2007.
 137.Moesgaard SG, Olsen LH, Viuff BM, Baandrup U, Pedersen LG, Thomsen PD, Pedersen HD, Harrison AP. Increased nitric oxide release and expression of endothelial and inducible nitric oxide synthases in mildly changed porcine mitral valve leaflets. J Heart Valve Dis 16: 67‐75, 2007.
 138.Mohler ER, III, Gannon F, Reynolds C, Zimmerman R, Keane MG, Kaplan FS. Bone formation and inflammation in cardiac valves. Circulation 103: 1522‐1528, 2001.
 139.Mokadam NA, Stout KK, Verrier ED. Management of acute regurgitation in left‐sided cardiac valves. Tex Heart Inst J 38: 9‐19, 2011.
 140.Morony S, Tintut Y, Zhang Z, Cattley RC, Van G, Dwyer D, Stolina M, Kostenuik PJ, Demer LL. Osteoprotegerin inhibits vascular calcification without affecting atherosclerosis in ldlr(−/−) mice. Circulation 117: 411‐420, 2008.
 141.Mow T, Poulsen K, Nielsen AH, Baandrup U, Hansen BF, Pedersen HD. Species differences in expression of angiotensin II receptors and angiotensin‐converting enzyme in human, canine and rat mitral valve leaflets. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 124: 11‐17, 1999.
 142.Natorska J, Marek G, Sadowski J, Undas A. Presence of B cells within aortic valves in patients with aortic stenosis: Relation to severity of the disease. J Cardiol 67: 80‐85, 2016.
 143.New SE, Aikawa E. Molecular imaging insights into early inflammatory stages of arterial and aortic valve calcification. Circ Res 108: 1381‐1391, 2011.
 144.Ngo MV, Gottdiener JS, Fletcher RD, Fernicola DJ, Gersh BJ. Smoking and obesity are associated with the progression of aortic stenosis. Am J Geriatr Cardiol 10: 86‐90, 2001.
 145.Ni CW, Qiu H, Rezvan A, Kwon K, Nam D, Son DJ, Visvader JE, Jo H. Discovery of novel mechanosensitive genes in vivo using mouse carotid artery endothelium exposed to disturbed flow. Blood 116: e66‐e73, 2010.
 146.Nigam V, Srivastava D. Notch1 represses osteogenic pathways in aortic valve cells. J Mol Cell Cardiol 47: 828‐834, 2009.
 147.Nishimura RA, Otto CM, Bonow RO, Carabello BA, Erwin JP, III, Guyton RA, O'Gara PT, Ruiz CE, Skubas NJ, Sorajja P, Sundt TM, III, Thomas JD, Anderson JL, Halperin JL, Albert NM, Bozkurt B, Brindis RG, Creager MA, Curtis LH, DeMets D, Guyton RA, Hochman JS, Kovacs RJ, Ohman EM, Pressler SJ, Sellke FW, Shen WK, Stevenson WG, Yancy CW, American College of C, American College of Cardiology/American Heart A, and American Heart A. 2014 AHA/ACC guideline for the management of patients with valvular heart disease: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Thorac Cardiovasc Surg 148: e1‐e132, 2014.
 148.Nkomo VT, Gardin JM, Skelton TN, Gottdiener JS, Scott CG, Enriquez‐Sarano M. Burden of valvular heart diseases: A population‐based study. Lancet 368: 1005‐1011, 2006.
 149.Obadia JF, Casali C, Chassignolle JF, Janier M. Mitral subvalvular apparatus: Different functions of primary and secondary chordae. Circulation 96: 3124‐3128, 1997.
 150.Obayashi K, Miyagawa‐Tomita S, Matsumoto H, Koyama H, Nakanishi T, Hirose H. Effects of transforming growth factor‐beta3 and matrix metalloproteinase‐3 on the pathogenesis of chronic mitral valvular disease in dogs. Am J Vet Res 72: 194‐202, 2011.
 151.Oliveira‐Paula GH, Lacchini R, Tanus‐Santos JE. Inducible nitric oxide synthase as a possible target in hypertension. Curr Drug Targets 15: 164‐174, 2014.
 152.Orita Y, Yamamoto H, Kohno N, Sugihara M, Honda H, Kawamata S, Mito S, Soe NN, Yoshizumi M. Role of osteoprotegerin in arterial calcification: Development of new animal model. Arterioscler Thromb Vasc Biol 27: 2058‐2064, 2007.
 153.Orton EC, Lacerda CM, MacLea HB. Signaling pathways in mitral valve degeneration. J Vet Cardiol 14: 7‐17, 2012.
 154.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 90: 844‐853, 1994.
 155.Oyama MA, Chittur SV. Genomic expression patterns of mitral valve tissues from dogs with degenerative mitral valve disease. Am J Vet Res 67: 1307‐1318, 2006.
 156.Oyama MA, Levy RJ. Insights into serotonin signaling mechanisms associated with canine degenerative mitral valve disease. J Vet Intern Med 24: 27‐36, 2010.
 157.Ozaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue N, Hirata K, Yasui H, Sakurai H, Yoshida Y, Masada M, Yokoyama M. Overexpression of endothelial nitric oxide synthase accelerates atherosclerotic lesion formation in apoE‐deficient mice. J Clin Invest 110: 331‐340, 2002.
 158.Palta S, Pai AM, Gill KS, Pai RG. New insights into the progression of aortic stenosis: Implications for secondary prevention. Circulation 101: 2497‐2502, 2000.
 159.Panizo S, Cardus A, Encinas M, Parisi E, Valcheva P, Lopez‐Ongil S, Coll B, Fernandez E, Valdivielso JM. RANKL increases vascular smooth muscle cell calcification through a RANK‐BMP4‐dependent pathway. Circ Res 104: 1041‐1048, 2009.
 160.Paranya G, Vineberg S, Dvorin E, Kaushal S, Roth SJ, Rabkin E, Schoen FJ, Bischoff J. Aortic valve endothelial cells undergo transforming growth factor‐beta‐mediated and non‐transforming growth factor‐beta‐mediated transdifferentiation in vitro. Am J Pathol 159: 1335‐1343, 2001.
 161.Piera‐Velazquez S, Li Z, Jimenez SA. Role of endothelial‐mesenchymal transition (EndoMT) in the pathogenesis of fibrotic disorders. Am J Pathol 179: 1074‐1080, 2011.
 162.Poggio P, Branchetti E, Grau JB, Lai EK, Gorman RC, Gorman JH, III, Sacks MS, Bavaria JE, Ferrari G. Osteopontin‐CD44v6 interaction mediates calcium deposition via phospho‐Akt in valve interstitial cells from patients with noncalcified aortic valve sclerosis. Arterioscler Thromb Vasc Biol 34: 2086‐2094, 2014.
 163.Poggio P, Folesani G, Raffa GM, Songia P, Valenti V, Myasoedova V, Parolari A. Antihypertensive treatments in patients affected by aortic valve stenosis. Curr Pharm Des 2016.
 164.Poggio P, Sainger R, Branchetti E, Grau JB, Lai EK, Gorman RC, Sacks MS, Parolari A, Bavaria JE, Ferrari G. Noggin attenuates the osteogenic activation of human valve interstitial cells in aortic valve sclerosis. Cardiovasc Res 98: 402‐410, 2013.
 165.Pohjolainen V, Taskinen P, Soini Y, Rysa J, Ilves M, Juvonen T, Ruskoaho H, Leskinen H, Satta J. Noncollagenous bone matrix proteins as a part of calcific aortic valve disease regulation. Hum Pathol 39: 1695‐1701, 2008.
 166.Qin X, Corriere MA, Matrisian LM, Guzman RJ. Matrix metalloproteinase inhibition attenuates aortic calcification. Arterioscler Thromb Vasc Biol 26: 1510‐1516, 2006.
 167.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 104: 2525‐2532, 2001.
 168.Rajamannan NM. Mechanisms of aortic valve calcification: The LDL‐density‐radius theory: A translation from cell signaling to physiology. Am J Physiol Heart Circ Physiol 298: H5‐H15, 2010.
 169.Rajamannan NM, Evans FJ, Aikawa E, Grande‐Allen KJ, Demer LL, Heistad DD, Simmons CA, Masters KS, Mathieu P, O'Brien KD, Schoen FJ, Towler DA, Yoganathan AP, Otto CM. Calcific aortic valve disease: Not simply a degenerative process: A review and agenda for research from the National Heart and Lung and Blood Institute Aortic Stenosis Working Group. Executive summary: Calcific aortic valve disease‐2011 update. Circulation 124: 1783‐1791, 2011.
 170.Rajamannan NM, Subramaniam M, Caira F, Stock SR, Spelsberg TC. Atorvastatin inhibits hypercholesterolemia‐induced calcification in the aortic valves via the Lrp5 receptor pathway. Circulation 112: I229‐234, 2005.
 171.Rajamannan NM, Subramaniam M, Stock SR, Stone NJ, Springett M, Ignatiev KI, McConnell JP, Singh RJ, Bonow RO, Spelsberg TC. Atorvastatin inhibits calcification and enhances nitric oxide synthase production in the hypercholesterolaemic aortic valve. Heart 91: 806‐810, 2005.
 172.Ranganathan N, Lam JH, Wigle ED, Silver MD. Morphology of the human mitral valve. II. The value leaflets. Circulation 41: 459‐467, 1970.
 173.Rao TP, Kuhl M. An updated overview on Wnt signaling pathways: A prelude for more. Circ Res 106: 1798‐1806, 2010.
 174.Rapaport E. Natural history of aortic and mitral valve disease. Am J Cardiol 35: 221‐227, 1975.
 175.Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator‐activated receptor‐gamma is a negative regulator of macrophage activation. Nature 391: 79‐82, 1998.
 176.Ross J, Jr., Braunwald E. Aortic stenosis. Circulation 38: 61‐67, 1968.
 177.Rothman RB, Baumann MH. Serotonergic drugs and valvular heart disease. Expert Opin. Drug Saf 8: 317‐329, 2009.
 178.Rothman RB, Baumann MH, Savage JE, Rauser L, McBride A, Hufeisen SJ, Roth BL. Evidence for possible involvement of 5‐HT(2B) receptors in the cardiac valvulopathy associated with fenfluramine and other serotonergic medications. Circulation 102: 2836‐2841, 2000.
 179.Rusted IE, Scheifley CH, Edwards JE. Studies of the mitral valve. I. Anatomic features of the normal mitral valve and associated structures. Circulation 6: 825‐831, 1952.
 180.Sacks MS, David Merryman W, Schmidt DE. On the biomechanics of heart valve function. J Biomech 42: 1804‐1824, 2009.
 181.Sainger R, Grau JB, Branchetti E, Poggio P, Lai E, Koka E, Vernick WJ, Gorman RC, Bavaria JE, Ferrari G. Comparison of transesophageal echocardiographic analysis and circulating biomarker expression profile in calcific aortic valve disease. J Heart Valve Dis 22: 156‐165, 2013.
 182.Sainger R, Grau JB, Branchetti E, Poggio P, Seefried WF, Field BC, Acker MA, Gorman RC, Gorman JH, III, Hargrove CW, III, Bavaria JE, Ferrari G. Human myxomatous mitral valve prolapse: Role of bone morphogenetic protein 4 in valvular interstitial cell activation. J Cell Physiol 227: 2595‐2604, 2012.
 183.Sasaki H, Yamamoto H, Tominaga K, Masuda K, Kawai T, Teshima‐Kondo S, Matsuno K, Yabe‐Nishimura C, Rokutan K. Receptor activator of nuclear factor‐kappaB ligand‐induced mouse osteoclast differentiation is associated with switching between NADPH oxidase homologues. Free Radic Biol Med 47: 189‐199, 2009.
 184.Schade R, Andersohn F, Suissa S, Haverkamp W, Garbe E. Dopamine agonists and the risk of cardiac‐valve regurgitation. N Engl J Med 356: 29‐38, 2007.
 185.Schultz‐Cherry S, Murphy‐Ullrich JE. Thrombospondin causes activation of latent transforming growth factor‐beta secreted by endothelial cells by a novel mechanism. J Cell Biol 122: 923‐932, 1993.
 186.Scruggs SM, Disatian S, Orton EC. Serotonin transmembrane transporter is down‐regulated in late‐stage canine degenerative mitral valve disease. J Vet Cardiol 12: 163‐169, 2010.
 187.Seya K, Yu Z, Kanemaru K, Daitoku K, Akemoto Y, Shibuya H, Fukuda I, Okumura K, Motomura S, Furukawa K. Contribution of bone morphogenetic protein‐2 to aortic valve calcification in aged rat. J Pharmacol Sci 115: 8‐14, 2011.
 188.Siney L, Lewis MJ. Nitric oxide release from porcine mitral valves. Cardiovasc Res 27: 1657‐1661, 1993.
 189.Singh JP, Evans JC, Levy D, Larson MG, Freed LA, Fuller DL, Lehman B, Benjamin EJ. Prevalence and clinical determinants of mitral, tricuspid, and aortic regurgitation (the Framingham Heart Study). Am J Cardiol 83: 897‐902, 1999.
 190.Singh RG, Cappucci R, Kramer‐Fox R, Roman MJ, Kligfield P, Borer JS, Hochreiter C, Isom OW, Devereux RB. Severe mitral regurgitation due to mitral valve prolapse: Risk factors for development, progression, and need for mitral valve surgery. Am J Cardiol 85: 193‐198, 2000.
 191.Siu SC, Silversides CK. Bicuspid aortic valve disease. J Am Coll Cardiol 55: 2789‐2800, 2010.
 192.Songia P, Branchetti E, Parolari A, Myasoedova V, Ferrari G, Alamanni F, Tremoli E, Poggio P. Mitral valve endothelial cells secrete osteoprotegerin during endothelial mesenchymal transition. J Mol Cell Cardiol 98: 48‐57, 2016.
 193.Sorescu GP, Song H, Tressel SL, Hwang J, Dikalov S, Smith DA, Boyd NL, Platt MO, Lassegue B, Griendling KK, Jo H. Bone morphogenic protein 4 produced in endothelial cells by oscillatory shear stress induces monocyte adhesion by stimulating reactive oxygen species production from a nox1‐based NADPH oxidase. Circ Res 95: 773‐779, 2004.
 194.Stein PD, Wang CH, Riddle JM, Sabbah HN, Magilligan DJ, Jr., Hawkins ET. Scanning electron microscopy of operatively excised severely regurgitant floppy mitral valves. Am J Cardiol 64: 392‐394, 1989.
 195.Stephens EH, Nguyen TC, Itoh A, Ingels NB, Jr., Miller DC, Grande‐Allen KJ. The effects of mitral regurgitation alone are sufficient for leaflet remodeling. Circulation 118: S243‐S249, 2008.
 196.Sucosky P, Balachandran K, Elhammali A, Jo H, Yoganathan AP. Altered shear stress stimulates upregulation of endothelial VCAM‐1 and ICAM‐1 in a BMP‐4‐ and TGF‐beta1‐dependent pathway. Arterioscler Thromb Vasc Biol 29: 254‐260, 2009.
 197.Tanikawa T, Okada Y, Tanikawa R, Tanaka Y. Advanced glycation end products induce calcification of vascular smooth muscle cells through RAGE/p38 MAPK. J Vasc Res 46: 572‐580, 2009.
 198.Timek TA, Green GR, Tibayan FA, Lai DT, Rodriguez F, Liang D, Daughters GT, Ingels NB, Jr., Miller DC. Aorto‐mitral annular dynamics. Ann Thorac Surg 76: 1944‐1950, 2003.
 199.Timmerman LA, Grego‐Bessa J, Raya A, Bertran E, Perez‐Pomares JM, Diez J, Aranda S, Palomo S, McCormick F, Izpisua‐Belmonte JC, de la Pompa JL. Notch promotes epithelial‐mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev 18: 99‐115, 2004.
 200.Vahanian A, Otto CM. Risk stratification of patients with aortic stenosis. Eur Heart J 31: 416‐423, 2010.
 201.Vattikuti R, Towler DA. Osteogenic regulation of vascular calcification: An early perspective. Am J Physiol Endocrinol Metab 286: E686‐E696, 2004.
 202.Villar AV, Cobo M, Llano M, Montalvo C, Gonzalez‐Vilchez F, Martin‐Duran R, Hurle MA, Nistal JF. Plasma levels of transforming growth factor‐beta1 reflect left ventricular remodeling in aortic stenosis. PloS One 4: e8476, 2009.
 203.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 95: 253‐260, 2004.
 204.Waltenberger J, Lundin L, Oberg K, Wilander E, Miyazono K, Heldin CH, Funa K. Involvement of transforming growth factor‐beta in the formation of fibrotic lesions in carcinoid heart disease. Am J Pathol 142: 71‐78, 1993.
 205.Walther DJ, Peter JU, Bashammakh S, Hortnagl H, Voits M, Fink H, Bader M. Synthesis of serotonin by a second tryptophan hydroxylase isoform. Science 299: 76, 2003.
 206.Wipff PJ, Rifkin DB, Meister JJ, Hinz B. Myofibroblast contraction activates latent TGF‐beta1 from the extracellular matrix. J Cell Biol 179: 1311‐1323, 2007.
 207.Wirrig EE, Hinton RB, Yutzey KE. Differential expression of cartilage and bone‐related proteins in pediatric and adult diseased aortic valves. J Mol Cell Cardiol 50: 561‐569, 2011.
 208.Wu HD, Maurer MS, Friedman RA, Marboe CC, Ruiz‐Vazquez EM, Ramakrishnan R, Schwartz A, Tilson MD, Stewart AS, Winchester R. The lymphocytic infiltration in calcific aortic stenosis predominantly consists of clonally expanded T cells. J Immunol 178: 5329‐5339, 2007.
 209.Wylie‐Sears J, Aikawa E, Levine RA, Yang JH, Bischoff J. Mitral valve endothelial cells with osteogenic differentiation potential. Arterioscler Thromb Vasc Biol 31: 598‐607, 2011.
 210.Xu J, Jian B, Chu R, Lu Z, Li Q, Dunlop J, Rosenzweig‐Lipson S, McGonigle P, Levy RJ, Liang B. Serotonin mechanisms in heart valve disease II: The 5‐HT2 receptor and its signaling pathway in aortic valve interstitial cells. Am J Pathol 161: 2209‐2218, 2002.
 211.Yadav VK, Ducy P. Lrp5 and bone formation: A serotonin‐dependent pathway. Ann N Y Acad Sci 1192: 103‐109, 2010.
 212.Yamashita A, Takada T, Nemoto K, Yamamoto G, Torii R. Transient suppression of PPARgamma directed ES cells into an osteoblastic lineage. FEBS Lett 580: 4121‐4125, 2006.
 213.Yetkin E, Tchaikovski V, Erdil N, Alan S, Waltenberger J. Increased expression of cystatin C and transforming growth factor beta‐1 in calcific aortic valves. Int J Cardiol 176: 1252‐1254, 2014.
 214.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 29: 936‐942, 2009.
 215.Yoon BS, Lyons KM. Multiple functions of BMPs in chondrogenesis. J Cell Biochem 93: 93‐103, 2004.
 216.Yosefy C, Ben Barak A. Floppy mitral valve/mitral valve prolapse and genetics. J Heart Valve Dis 16: 590‐595, 2007.
 217.Yperman J, De Visscher G, Holvoet P, Flameng W. Molecular and functional characterization of ovine cardiac valve‐derived interstitial cells in primary isolates and cultures. Tissue Eng 10: 1368‐1375, 2004.
 218.Zanettini R, Antonini A, Gatto G, Gentile R, Tesei S, Pezzoli G. Valvular heart disease and the use of dopamine agonists for Parkinson's disease. N Engl J Med 356: 39‐46, 2007.

Teaching Material

G. L. Perrucci, M. Zanobini, P. Gripari, P. Songia, B. Alshaikh, E. Tremoli, P. Poggio. Pathophysiology of Aortic Stenosis and Mitral Regurgitation. Compr Physiol 7 2017, 799-818.

Didactic Synopsis

 

Major Teaching Points:

     

  1. Understanding the anatomical structures of aortic and mitral valves is necessary to apprehend the pathophysiology of aortic valve stenosis and mitral valve regurgitation.
  2.  

     

  3. The major pathological processes involved in aortic valve stenosis are:

       

    1. valve interstitial cell phenotypic switching in osteoblast-like cells.
    2.  

       

    3. endothelial-to-mesenchymal transition of valve endothelial cells.
    4.  

       

    5. inflammatory cell infiltration.
    6.  

       

    7. fibrosis and osteogenesis.
    8.  

  4.  

     

  5. The main cardiac pathologies caused by aortic valve stenosis are:

       

    1. left ventricle pressure overload.
    2.  

       

    3. cardiac hypertrophy.
    4.  

       

    5. syncope.
    6.  

       

    7. pulmonary hypertension.
    8.  

       

    9. angina.
    10.  

       

    11. heart failure.
    12.  

  6.  

     

  7. To date, the aortic valve stenosis management is represented by surgical intervention, since no pharmacological treatments have been approved.
  8.  

     

  9. The major pathological stimuli involved in mitral valve regurgitation are:

       

    1. mechanical such as are tension, shear stress, compression, and flexure.
    2.  

       

    3. chemical such as serotonin and TGF-β signaling.
    4.  

  10.  

     

  11. The main cardiac pathologies caused by mitral valve regurgitation are:

       

    1. left atrium pressure overload.
    2.  

       

    3. pulmonary edema.
    4.  

       

    5. pulmonary hypertension.
    6.  

       

    7. left ventricle dysfunction.
    8.  

       

    9. heart failure.
    10.  

  12.  

To date, mortality rates are extremely high without surgical intervention. However, patient with acute MR will also benefit from hemodynamically stabilization by afterload reduction accomplished either by pharmacological (e.g., vasodilators) or by mechanical treatments (e.g., intra-aortic balloon pump).

 

 

Didactic Legends

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

 

Figure 1. Teaching points: The aortic valve is an anatomical gate between the left ventricle and the ascending aorta. The aortic root components are: the leaflets, the junction of the aortic root with the ascending aorta, and the junction of the aortic root with the left ventricle.

Figure 2. Teaching points: Stenotic aortic valves show an impaired opening pattern, in systole, in comparison with healthy aortic valves. In physiological conditions, normal aortic valve leaflets are very thin, while in calcified aortic valves the leaflets show thickened edges.

Figure 3. Teaching points: The main cellular mechanisms involved in the pathogenesis of aortic valve stenosis are represented by the differentiation of monocytes into osteoclasts, by the endothelial damage leading to endothelial-to-mesenchymal transition of valve endothelial cells into activated myofibroblasts, by the differentiation of valve interstitial cells into activated myofibroblast and / or osteoblast-like cells, leading to fibrosis (such as collagen and proteoglycan deposition) and calcium nodules formation, respectively.

Figure 4. Teaching points: the stenotic valve shows several regions of spotty calcification. The stiffness of the valve impedes the correct opening of the leaflets during systole. The calcification of the aortic valve leads to left ventricle pressure overload, cardiac hypertrophy, syncope, pulmonary hypertension, heart failure, and angina.

Figure 5. Teaching points: The mitral valve is an anatomical gate between left atrium and left ventricle. The opening of mitral valve is controlled by cardiac papillary muscle contraction. The chordae tendinae linked to the papillary muscles guide the closing of the leaflets during systole.

Figure 6. Teaching points: Healthy mitral valves present a correct alignment of valve leaflets, while, the prolapsed one show an impaired closure. Clinically, the blood regurgitation caused by impaired closure of prolapsed mitral leaflets could be visualized by color-Doppler flow imaging.

Figure 7. Teaching points: The main cellular mechanisms involved in the pathogenesis of mitral valve regurgitation are represented by the endothelial damage leading to endothelial-to-mesenchymal transition of valve endothelial cells into activated myofibroblasts, by the fragmentation of elastin in the atrialis layer, by the differentiation of valve interstitial cells into activated myofibroblasts, leading to fibrosis (collagen and glycosaminoglycans deposition in the spongiosa layer), and by the collagen fragmentation in the fibrosa layer.

Figure 8. Teaching points: The regurgitant mitral valve improperly close at the end of systole. Pathologies caused by mitral valve regurgitation lead to left atrium pressure overload, pulmonary edema, pulmonary hypertension, left ventricle dysfunction, and heart failure.

 


Related Articles:

Cardiac Fibroblast Physiology and Pathology
Cardiac Physiology of Aging: Extracellular Considerations
Mechanisms of Cardiac Pain
Aging Effects on Cardiac Progenitor Cell Physiology
Cellular Basis of Physiological and Pathological Myocardial Growth

Contact Editor

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

* Required Field

How to Cite

Gianluca L Perrucci, Marco Zanobini, Paola Gripari, Paola Songia, Bayan Alshaikh, Elena Tremoli, Paolo Poggio. Pathophysiology of Aortic Stenosis and Mitral Regurgitation. Compr Physiol 2017, 7: 799-818. doi: 10.1002/cphy.c160020