Comprehensive Physiology Wiley Online Library

Heart Valve Biomechanics and Underlying Mechanobiology

Full Article on Wiley Online Library



ABSTRACT

Heart valves control unidirectional blood flow within the heart during the cardiac cycle. They have a remarkable ability to withstand the demanding mechanical environment of the heart, achieving lifetime durability by processes involving the ongoing remodeling of the extracellular matrix. The focus of this review is on heart valve functional physiology, with insights into the link between disease‐induced alterations in valve geometry, tissue stress, and the subsequent cell mechanobiological responses and tissue remodeling. We begin with an overview of the fundamentals of heart valve physiology and the characteristics and functions of valve interstitial cells (VICs). We then provide an overview of current experimental and computational approaches that connect VIC mechanobiological response to organ‐ and tissue‐level deformations and improve our understanding of the underlying functional physiology of heart valves. We conclude with a summary of future trends and offer an outlook for the future of heart valve mechanobiology, specifically, multiscale modeling approaches, and the potential directions and possible challenges of research development. © 2016 American Physiological Society. Compr Physiol 6:1743‐1780, 2016.

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. A schematic approach to the biomechanics and mechanobiology of heart valve function. Heart valve function can be divided into multiple length scales: organ, tissue, cell, and subcell levels.
Figure 2. Figure 2. The multiscale nature of heart valve biomechanics: a representation of the MV at the organ, tissue, and cell levels. At the tissue level: a circumferentially oriented cross section of the MV anterior leaflet stained with Movat pentachrome, which colors collagen yellow, elastic fibers black, and hydrated PGs and GAGs blue. At the cell‐level: a transmission electron micrograph of a mitral VIC from the fibrosa layer.
Figure 3. Figure 3. Scanning electron micrograph of the multilayered microenvironment of the MVAL. Individual micrographs of each layer are also presented: elastin‐rich ventricularis and atrialis, highly collagenous fibrosa, and proteoglycan‐rich spongiosa. The collagen fibrils and elastic fibers closely surround the interstitial cells and highlight the long cellular extensions. In the fibrosa, collagen fibrils are aligned in the circumferential direction of the leaflet, which is responsible for the observed anisotropy in leaflet mechanical behavior. (T: transmural, C: circumferential.)
Figure 4. Figure 4. A conceptual framework of the natural history of CAVD. The spectrum of disease ranges from the “at risk” patient to the patient with end‐stage severe symptomatic aortic stenosis. The light blue line represents the expected event‐free survival and the purple line represents the survival curve at the onset of aortic sclerosis, which deviates from the expected survival line. Atherosclerotic risk factors involved in the development of AV disease from aortic sclerosis to calcified AV disease are included. Adapted from Otto et al. (184) with permission and Rajamannan et al. (205).
Figure 5. Figure 5. Numerical simulation of the unsteady, pulsatile flow in a tricuspid prosthetic HV. Contours of the out‐of‐plane vorticity are shown at two phases during the cardiac cycle: (A) fully open phase and (B) closing phase.
Figure 6. Figure 6. Instantaneous friction streamline and shear stress magnitude plots on the aortic (A and C) and ventricular (B and D) sides of the leaflets during the fully open (A and B) and early closing (C and D) phases of the cardiac cycle.
Figure 7. Figure 7. (A) Schematic of the MVAL with the nine transducers used for sonomicrometry array localization. (B) Representative areal strain traces and corresponding areal strain rate data. The high strain rates (on the order of 1000% s−1) underscore the highly dynamic nature of the MV. Once the valve coapts fully, no further deformations occurred. (C) Mean principal strains for three pressure levels: 90, 150, and 200 mmHg. Other than the differences between the circumferential and radial peak strains, there were no significant differences in strain with increasing LV pressure. Adapted, with permission, from Sacks et al. (216).
Figure 8. Figure 8. (A) Diagram of the AV cusp highlighting the belly, commissures, nodulus, and regions of coaptation. SALS results with the orientation index at (B) 0mmHg, (C) 4 mmHg, and (D) 90 mmHg TVP. No further changes in fiber alignment were observed past 4 mmHg. These results are consistent with histological‐based data that quantifies the percent area of tissue displaying collagen fiber crimp (E). Adapted, with permission, from Sacks et al. (219).
Figure 9. Figure 9. Movat pentachrome stain of the transverse‐radial section of the center region of the MVAL, and multiphoton microscopy of the ventricularis, fibrosa, spongiosa, and atrialis layer of the anterior leaflet is also shown. Adapted, with permission, from Zhang et al. (302).
Figure 10. Figure 10. The net stress contribution from each ECM component from each layer for the equibiaxial stress protocol is shown for the circumferential and radial direction of the anterior and posterior leaflets. Here, the contributions from the layers are ventricularis (V), fibrosa (F), spongiosa (S), and atrialis (A). Interestingly, while the fibrosa layer is dominant circumferential directions in both leaflets, the atrialis also contributes substantially in the radial direction. Adapted, with permission, from Zhang et al. (302).
Figure 11. Figure 11. (A) The circumferential and radial stretches of the MV leaflet at the 90 N/m equitension state, λC peak and λR peak, revealed no significant differences among the prescribed set of loading time protocols in both the circumferential (P = 0.987) and radial (P = 0.996) directions. Stretches observed previously in mock flow loop (solid horizontal lines) ±SEM (dotted lines) are plotted for the circumferential (black) and radial (gray) specimen axes. λC peak and λR peakexceeded those observed in vitro; however, the ratio of λC peak to λR peak (0.86 ± 0.02) was very close to the ratio of peak circumferential and radial stretches observed in vitro (0.83). (B) Representative biaxial stress‐relaxation data, demonstrating continued relaxation throughout the 3‐h time frame. (C) Representative stretch versus time curves for a typical biaxial creep experiment. Adapted, with permission, from Zhang et al. (302).
Figure 12. Figure 12. (A) Schematic showing the region of tissue used for bending tests in the AV. (B) Schematic showing directions of bending for the AV with respective layers (V: ventricularis, S: spongiosa, and F: fibrosa). M versus Dk relations in both the AC and WC directions for (C) specimens tested in 5 and 90 mmol/L KCl, and (D) specimens flexed in 5 mmol/L KCl and samples treated in 10 mmol/L thapsigargin overnight and then flexed in 5 mmol/L KCl. While the application of 90 mmol/L KCL induced an increase in stiffness in the AC direction only, both bending directions experienced a loss of stiffness with the addition of thapsigargin to the bathing medium.
Figure 13. Figure 13. (A) The relation between AVIC NAR and TVP loading, with values reported over normalized leaflet thickness. (B) AVIC NAR versus the normalized collagen fiber orientation index (NOI) at different TVP levels. These results suggest that AVICs are not appreciably loaded until the collagen fibers fully straighten at TVPs more than ∼4 mmHg.
Figure 14. Figure 14. Transmission electron micrograph of an MVIC from the fibrosa layer of the MV anterior leaflet highlighting the cellular microenvironment, particularly collagen fibril circumferential orientation and close interaction and alignment with the MVIC. Scale bar, 1 μm. (T: transmural, R: radial, C: circumferential.)
Figure 15. Figure 15. (A) A VIC under MA—the vertical bar represents the aspiration length. (B) Functional correlations of effective cell stiffness E versus TVP. (C) Linear correlation between Hsp47 versus α‐SMA, showing a strong correlation between the two proteins (r = 0.996) as the one progresses from the right to the left side of the heart.
Figure 16. Figure 16. (A and B) Transmural MVIC distribution and deformation in the MV anterior leaflet under controlled biaxial loading. (A) Stack of two‐photon excited fluorescence images. Red represents collagen fibers and green represents cell nuclei stained with Cytox Green. Image stacks were processed and used to quantify cellular deformation by measuring the NAR. (B) Measured layer‐specific NAR as a function of membrane tension. (A: atrialis, S: spongiosa, F: fibrosa, V: ventricularis.) (C and D) The macro‐micro finite element (FE) model used to investigate MVIC deformation. (C) Schematic diagram of the tissue‐level model: the region of interest of the MVAL tissue under equibiaxial tension loading. (D) Diagram of the cell‐level microenvironment model, which consists of 37 uniformly distributed MVICs embedded in the layer‐specific representative volume element. The tissue‐level deformations are used to prescribe boundary displacements. Adapted, with permission, from Lee et al. (134).
Figure 17. Figure 17. Comparisons of the experimentally measured and numerically predicted NARs as a function of tissue‐level tension: (A) atrialis layer (r2 = 0.8403), (B) spongiosa layer (r2 = 0.9166), (C) fibrosa layer (r2 = 0.8906), and (D) ventricularis layer (r2 = 0.9373). Adapted, with permission, from Lee et al. (134).
Figure 18. Figure 18. AFM simulation setup with finite element mesh. (A) The volume ratio of the nucleus was acquired from electron micrographs of in situ VICs. Scale bar = 1 μm. VIC dimensions were acquired from height maps from AFM experiments. The colors represent the height of each point in μm. Scale bar = 10 μm. (B) The mesh was refined around the indentation region, with typically 10,000∼50,000 linear tetrahedron elements.
Figure 19. Figure 19. (A) Deformation contours central and peripheral indentations highlight that VIC deformation is localized around the indentation region. The simulations show that indentations in the central region deform the nucleus more than those at the periphery. (B) Simulated and experimental indentation data at the center can be used to analyze VIC nucleus mechanical properties.
Figure 20. Figure 20. The step‐by‐step calibration process. (A) The cytoskeletal shear modulus (μcyto) and α‐SMA fiber shear modulus (μsf) were calibrated using the MA data for different types of VICs with different expression levels of α‐SMA fibers. (B) Using the AFM indentation data on the cell peripheral regions, the fiber active contraction strength (f) was calibrated for AVICs and PVICs. (C) Using the AFM indentation data on the cell central region, the shear modulus of nucleus (μnuc) was calibrated for AVICs and PVICs. The parameters in red represents the ones that are being calibrated in the step, the parameters in gray represents the ones that are assumed not contribute to the mechanical response of the VICs (hence ignored) in the step, and the parameters in black represents the ones that are already calibrated from the previous steps and integrated into the model. Adapted, with permission, from Sakamoto et al. (223).
Figure 21. Figure 21. (A) Contraction strength of the α‐SMA fibers in the AVICs and PVICs. About 9:1 ratio in the contraction strength was observed from AVICs to PVICs. (B) Shear moduli of the AVICs and PVICs nuclei, which exhibited no differences. Adapted, with permission, from Sakamoto et al. (223).


Figure 1. A schematic approach to the biomechanics and mechanobiology of heart valve function. Heart valve function can be divided into multiple length scales: organ, tissue, cell, and subcell levels.


Figure 2. The multiscale nature of heart valve biomechanics: a representation of the MV at the organ, tissue, and cell levels. At the tissue level: a circumferentially oriented cross section of the MV anterior leaflet stained with Movat pentachrome, which colors collagen yellow, elastic fibers black, and hydrated PGs and GAGs blue. At the cell‐level: a transmission electron micrograph of a mitral VIC from the fibrosa layer.


Figure 3. Scanning electron micrograph of the multilayered microenvironment of the MVAL. Individual micrographs of each layer are also presented: elastin‐rich ventricularis and atrialis, highly collagenous fibrosa, and proteoglycan‐rich spongiosa. The collagen fibrils and elastic fibers closely surround the interstitial cells and highlight the long cellular extensions. In the fibrosa, collagen fibrils are aligned in the circumferential direction of the leaflet, which is responsible for the observed anisotropy in leaflet mechanical behavior. (T: transmural, C: circumferential.)


Figure 4. A conceptual framework of the natural history of CAVD. The spectrum of disease ranges from the “at risk” patient to the patient with end‐stage severe symptomatic aortic stenosis. The light blue line represents the expected event‐free survival and the purple line represents the survival curve at the onset of aortic sclerosis, which deviates from the expected survival line. Atherosclerotic risk factors involved in the development of AV disease from aortic sclerosis to calcified AV disease are included. Adapted from Otto et al. (184) with permission and Rajamannan et al. (205).


Figure 5. Numerical simulation of the unsteady, pulsatile flow in a tricuspid prosthetic HV. Contours of the out‐of‐plane vorticity are shown at two phases during the cardiac cycle: (A) fully open phase and (B) closing phase.


Figure 6. Instantaneous friction streamline and shear stress magnitude plots on the aortic (A and C) and ventricular (B and D) sides of the leaflets during the fully open (A and B) and early closing (C and D) phases of the cardiac cycle.


Figure 7. (A) Schematic of the MVAL with the nine transducers used for sonomicrometry array localization. (B) Representative areal strain traces and corresponding areal strain rate data. The high strain rates (on the order of 1000% s−1) underscore the highly dynamic nature of the MV. Once the valve coapts fully, no further deformations occurred. (C) Mean principal strains for three pressure levels: 90, 150, and 200 mmHg. Other than the differences between the circumferential and radial peak strains, there were no significant differences in strain with increasing LV pressure. Adapted, with permission, from Sacks et al. (216).


Figure 8. (A) Diagram of the AV cusp highlighting the belly, commissures, nodulus, and regions of coaptation. SALS results with the orientation index at (B) 0mmHg, (C) 4 mmHg, and (D) 90 mmHg TVP. No further changes in fiber alignment were observed past 4 mmHg. These results are consistent with histological‐based data that quantifies the percent area of tissue displaying collagen fiber crimp (E). Adapted, with permission, from Sacks et al. (219).


Figure 9. Movat pentachrome stain of the transverse‐radial section of the center region of the MVAL, and multiphoton microscopy of the ventricularis, fibrosa, spongiosa, and atrialis layer of the anterior leaflet is also shown. Adapted, with permission, from Zhang et al. (302).


Figure 10. The net stress contribution from each ECM component from each layer for the equibiaxial stress protocol is shown for the circumferential and radial direction of the anterior and posterior leaflets. Here, the contributions from the layers are ventricularis (V), fibrosa (F), spongiosa (S), and atrialis (A). Interestingly, while the fibrosa layer is dominant circumferential directions in both leaflets, the atrialis also contributes substantially in the radial direction. Adapted, with permission, from Zhang et al. (302).


Figure 11. (A) The circumferential and radial stretches of the MV leaflet at the 90 N/m equitension state, λC peak and λR peak, revealed no significant differences among the prescribed set of loading time protocols in both the circumferential (P = 0.987) and radial (P = 0.996) directions. Stretches observed previously in mock flow loop (solid horizontal lines) ±SEM (dotted lines) are plotted for the circumferential (black) and radial (gray) specimen axes. λC peak and λR peakexceeded those observed in vitro; however, the ratio of λC peak to λR peak (0.86 ± 0.02) was very close to the ratio of peak circumferential and radial stretches observed in vitro (0.83). (B) Representative biaxial stress‐relaxation data, demonstrating continued relaxation throughout the 3‐h time frame. (C) Representative stretch versus time curves for a typical biaxial creep experiment. Adapted, with permission, from Zhang et al. (302).


Figure 12. (A) Schematic showing the region of tissue used for bending tests in the AV. (B) Schematic showing directions of bending for the AV with respective layers (V: ventricularis, S: spongiosa, and F: fibrosa). M versus Dk relations in both the AC and WC directions for (C) specimens tested in 5 and 90 mmol/L KCl, and (D) specimens flexed in 5 mmol/L KCl and samples treated in 10 mmol/L thapsigargin overnight and then flexed in 5 mmol/L KCl. While the application of 90 mmol/L KCL induced an increase in stiffness in the AC direction only, both bending directions experienced a loss of stiffness with the addition of thapsigargin to the bathing medium.


Figure 13. (A) The relation between AVIC NAR and TVP loading, with values reported over normalized leaflet thickness. (B) AVIC NAR versus the normalized collagen fiber orientation index (NOI) at different TVP levels. These results suggest that AVICs are not appreciably loaded until the collagen fibers fully straighten at TVPs more than ∼4 mmHg.


Figure 14. Transmission electron micrograph of an MVIC from the fibrosa layer of the MV anterior leaflet highlighting the cellular microenvironment, particularly collagen fibril circumferential orientation and close interaction and alignment with the MVIC. Scale bar, 1 μm. (T: transmural, R: radial, C: circumferential.)


Figure 15. (A) A VIC under MA—the vertical bar represents the aspiration length. (B) Functional correlations of effective cell stiffness E versus TVP. (C) Linear correlation between Hsp47 versus α‐SMA, showing a strong correlation between the two proteins (r = 0.996) as the one progresses from the right to the left side of the heart.


Figure 16. (A and B) Transmural MVIC distribution and deformation in the MV anterior leaflet under controlled biaxial loading. (A) Stack of two‐photon excited fluorescence images. Red represents collagen fibers and green represents cell nuclei stained with Cytox Green. Image stacks were processed and used to quantify cellular deformation by measuring the NAR. (B) Measured layer‐specific NAR as a function of membrane tension. (A: atrialis, S: spongiosa, F: fibrosa, V: ventricularis.) (C and D) The macro‐micro finite element (FE) model used to investigate MVIC deformation. (C) Schematic diagram of the tissue‐level model: the region of interest of the MVAL tissue under equibiaxial tension loading. (D) Diagram of the cell‐level microenvironment model, which consists of 37 uniformly distributed MVICs embedded in the layer‐specific representative volume element. The tissue‐level deformations are used to prescribe boundary displacements. Adapted, with permission, from Lee et al. (134).


Figure 17. Comparisons of the experimentally measured and numerically predicted NARs as a function of tissue‐level tension: (A) atrialis layer (r2 = 0.8403), (B) spongiosa layer (r2 = 0.9166), (C) fibrosa layer (r2 = 0.8906), and (D) ventricularis layer (r2 = 0.9373). Adapted, with permission, from Lee et al. (134).


Figure 18. AFM simulation setup with finite element mesh. (A) The volume ratio of the nucleus was acquired from electron micrographs of in situ VICs. Scale bar = 1 μm. VIC dimensions were acquired from height maps from AFM experiments. The colors represent the height of each point in μm. Scale bar = 10 μm. (B) The mesh was refined around the indentation region, with typically 10,000∼50,000 linear tetrahedron elements.


Figure 19. (A) Deformation contours central and peripheral indentations highlight that VIC deformation is localized around the indentation region. The simulations show that indentations in the central region deform the nucleus more than those at the periphery. (B) Simulated and experimental indentation data at the center can be used to analyze VIC nucleus mechanical properties.


Figure 20. The step‐by‐step calibration process. (A) The cytoskeletal shear modulus (μcyto) and α‐SMA fiber shear modulus (μsf) were calibrated using the MA data for different types of VICs with different expression levels of α‐SMA fibers. (B) Using the AFM indentation data on the cell peripheral regions, the fiber active contraction strength (f) was calibrated for AVICs and PVICs. (C) Using the AFM indentation data on the cell central region, the shear modulus of nucleus (μnuc) was calibrated for AVICs and PVICs. The parameters in red represents the ones that are being calibrated in the step, the parameters in gray represents the ones that are assumed not contribute to the mechanical response of the VICs (hence ignored) in the step, and the parameters in black represents the ones that are already calibrated from the previous steps and integrated into the model. Adapted, with permission, from Sakamoto et al. (223).


Figure 21. (A) Contraction strength of the α‐SMA fibers in the AVICs and PVICs. About 9:1 ratio in the contraction strength was observed from AVICs to PVICs. (B) Shear moduli of the AVICs and PVICs nuclei, which exhibited no differences. Adapted, with permission, from Sakamoto et al. (223).
References
 1.Abrams J. The aortic valve by Mano Thubrikar Crc Press, Inc., Boca Raton (1990) 221 pages, illustrated, $97.50 ISBN: 0–8493–4771–8. Clin Cardiol 14: 364a‐365, 1991.
 2.Accola KD, Scott ML, Thompson PA, Palmer GJ, III, Sand ME, Ebra G. Midterm outcomes using the physio ring in mitral valve reconstruction: Experience in 492 patients. Ann Thorac Surg 79: 1276‐1283; discussion 1276‐1283, 2005.
 3.Adamczyk MM, Vesely I. Characteristics of compressive strains in porcine aortic valves cusps. J Heart Valve Dis 11: 75‐83, 2002.
 4.Aikawa E, Otto CM. Look more closely at the valve: Imaging calcific aortic valve disease. Circulation 125: 9‐11, 2012.
 5.Aikawa E, Whittaker P, Farber M, Mendelson K, Padera RF, Aikawa M, Schoen FJ. Human semilunar cardiac valve remodeling by activated cells from fetus to adult: Implications for postnatal adaptation, pathology, and tissue engineering. Circulation 113: 1344‐1352, 2006.
 6.Akdemir R, Ozhan H, Bulur S, Unlu H, Gunduz H, Arinc H, Yildiz A, Uyan C. Color M‐mode regurgitant flow propagation velocity: A new echocardiographic method for grading of mitral regurgitation. Echocardiography (Mount Kisco, NY 22: 713‐722, 2005.
 7.Alfieri CM, Cheek J, Chakraborty S, Yutzey KE. Wnt signaling in heart valve development and osteogenic gene induction. Dev Biol 338: 127‐135, 2010.
 8.Anderson RH, Ho SY, Becker AE. Anatomy of the human atrioventricular junctions revisited. Anat Rec 260: 81‐91, 2000.
 9.Armstrong EJ, Bischoff J. Heart valve development: Endothelial cell signaling and differentiation. Circ Res 95: 459‐470, 2004.
 10.Arts T, Meerbaum S, Reneman R, Corday E. Stresses in the closed mitral valve: A model study. J Biomech 16: 539‐547, 1983.
 11.Bairati A, DeBiasi S. Presence of a smooth muscle system in aortic valve leaflets. Anat Embryol (Berl) 161: 329‐340, 1981.
 12.Balachandran K, Alford PW, Wylie‐Sears J, Goss JA, Grosberg A, Bischoff J, Aikawa E, Levine RA, Parker KK. Cyclic strain induces dual‐mode endothelial‐mesenchymal transformation of the cardiac valve. Proc Natl Acad Sci U S A 108: 19943‐19948, 2011.
 13.Balachandran K, Konduri S, Sucosky P, Jo H, Yoganathan A. An ex vivo study of the biological properties of porcine aortic valves in response to circumferential cyclic stretch. Ann Biomed Eng 34: 1655‐1665, 2006.
 14.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 296: H756‐H764, 2009.
 15.Barczyk M, Carracedo S, Gullberg D. Integrins. Cell Tissue Res 339: 269‐280, 2010.
 16.Beckmann E, Grau JB, Sainger R, Poggio P, Ferrari G. Insights into the use of biomarkers in calcific aortic valve disease. J Heart Valve Dis 19: 441‐452, 2010.
 17.Bellhouse BJ, Bellhouse FH. Fluid mechanics of the mitral valve. Nature 224: 615‐618, 1969.
 18.Bellhouse BJ, Reid KG. Fluid mechanics of the aortic valve. Br Heart J 31: 391, 1969.
 19.Benton JA, Kern HB, Leinwand LA, Mariner PD, Anseth KS. Statins block calcific nodule formation of valvular interstitial cells by inhibiting alpha‐smooth muscle actin expression. Arterioscler Thromb Vasc Biol 29: 1950‐1957, 2009.
 20.Beppu H, Malhotra R, Beppu Y, Lepore JJ, Parmacek MS, Bloch KD. BMP type II receptor regulates positioning of outflow tract and remodeling of atrioventricular cushion during cardiogenesis. Dev Biol 331: 167‐175, 2009.
 21.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 122: 327‐335, 2000b.
 22.Billiar KL, Sacks MS. Biaxial mechanical properties of the natural and glutaraldehyde treated aortic valve cusp–Part I: Experimental results. J Biomech Eng 122: 23‐30, 2000a.
 23.Blevins TL, Carroll JL, Raza AM, Grande‐Allen KJ. Phenotypic characterization of isolated valvular interstitial cell subpopulations. J Heart Valve Dis 15: 815‐822, 2006.
 24.Booz GW, Baker KM. Molecular signalling mechanisms controlling growth and function of cardiac fibroblasts. Cardiovasc Res 30: 537‐543, 1995.
 25.Bouma W, Lai EK, Levack MM, Shang EK, Pouch AM, Eperjesi TJ, Plappert TJ, Yushkevich PA, Mariani MA, Khabbaz KR, Gleason TG, Mahmood F, Acker MA, Woo YJ, Cheung AT, Jackson BM, Gorman JH, III, Gorman RC. Preoperative three‐dimensional valve analysis predicts recurrent ischemic mitral regurgitation after mitral annuloplasty. Ann Thorac Surg 101: 567‐575, 2016.
 26.Buchanan RM, Sacks MS. Interlayer micromechanics of the aortic heart valve leaflet. Biomech Model Mechanobiol 13: 813‐826, 2013.
 27.Butcher JT, Mahler GJ, Hockaday LA. Aortic valve disease and treatment: The need for naturally engineered solutions. Adv Drug Deliv Rev 63: 242‐268, 2011.
 28.Butcher JT, Nerem RM. Porcine aortic valve interstitial cells in three‐dimensional culture: Comparison of phenotype with aortic smooth muscle cells. J Heart Valve Dis 13: 478‐485; discussion 485‐476, 2004.
 29.Butcher JT, Nerem RM. Valvular endothelial cells and the mechanoregulation of valvular pathology. Philos Trans R Soc Lond B Biol Sci 362: 1445‐1457, 2007.
 30.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.
 31.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.
 32.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.
 33.Carpentier A. Cardiac valve surgery–the “French correction.” J Thorac Cardiovasc Surg 86: 323‐337, 1983.
 34.Carpentier A, Chauvaud S, Fabiani JN, Deloche A, Relland J, Lessana A, D'Allaines C, Blondeau P, Piwnica A, Dubost C. Reconstructive surgery of mitral valve incompetence: Ten‐year appraisal. J Thorac Cardiovasc Surg 79: 338‐348, 1980.
 35.Carruthers CA, Good B, D'Amore A, Liao J, Amini R, Watkins SC, Sacks MS. Alterations in the microstructure of the anterior mitral valve leaflet under physiological stress. ASME 2012 Summer Bioengineering Conference. Fajardo, Puerto Rico: American Society of Mechanical Engineers, 2012, pp. 227‐228.
 36.Chakraborty S, Cheek J, Sakthivel B, Aronow BJ, Yutzey KE. Shared gene expression profiles in developing heart valves and osteoblast progenitor cells. Physiol Genomics 35: 75‐85, 2008.
 37.Chappell DC, Varner SE, Nerem RM, Medford RM, Alexander RW. Oscillatory shear stress stimulates adhesion molecule expression in cultured human endothelium. Circ Res 82: 532‐539, 1998.
 38.Chaput M, Handschumacher MD, Guerrero JL, Holmvang G, Dal‐Bianco JP, Sullivan S, Vlahakes GJ, Hung J, Levine RA. Mitral leaflet adaptation to ventricular remodeling: Prospective changes in a model of ischemic mitral regurgitation. Circulation 120: S99‐S103, 2009.
 39.Chaput M, Handschumacher MD, Tournoux F, Hua L, Guerrero JL, Vlahakes GJ, Levine RA. Mitral leaflet adaptation to ventricular remodeling: Occurrence and adequacy in patients with functional mitral regurgitation. Circulation 118: 845‐852, 2008.
 40.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.
 41.Chester AH, Kershaw JDB, Misfeld M, Sievers H‐H, Yacoub MH. Specific regional and direcrtional contractile response of aortic cusp tissue‐Relevance to valve function. Second Biennial Meeting of the Society for Heart Valve Disease, Paris, 2003, p. 67.
 42.Chester AH, Misfeld M, Yacoub MH. Receptor‐mediated contraction of aortic valve leaflets. J Heart Valve Dis 9: 250‐254; discussion 254‐255, 2000.
 43.Christie GW, Barratt‐Boyes BG. Age‐dependent changes in the radial stretch of human aortic valve leaflets determined by biaxial stretching. Ann Thorac Surg 60: S156‐S159, 1995.
 44.Christie GW, Barratt‐Boyes BG. Biaxial mechanical properties of explanted aortic allograft leaflets. Ann Thorac Surg 60: S160‐S164, 1995.
 45.Christie GW, Barratt‐Boyes BG. Mechanical properties of porcine pulmonary valve leaflets: How do they differ from aortic leaflets? Ann Thorac Surg 60: S195‐S199, 1995.
 46.Chuong CJ, Fung YC. On residual stress in arteries. J Biomech Eng 108: 189‐192, 1986.
 47.Cole WG, Chan D, Hickey AJ, Wilcken DE. Collagen composition of normal and myxomatous human mitral heart valves. Biochem J 219: 451‐460, 1984.
 48.Combs MD, Yutzey KE. Heart valve development: Regulatory networks in development and disease. Circ Res 105: 408‐421, 2009.
 49.Combs MD, Yutzey KE. VEGF and RANKL regulation of NFATc1 in heart valve development. Circ Res 105: 565‐574, 2009.
 50.Courtney T, Sacks MS, Stankus J, Guan J, Wagner WR. Design and analysis of tissue engineering scaffolds that mimic soft tissue mechanical anisotropy. Biomaterials 27: 3631‐3638, 2006.
 51.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.
 52.Cucina A, Sterpetti AV, Pupelis G, Fragale A, Lepidi S, Cavallaro A, Giustiniani Q, Santoro D'Angelo L. Shear stress induces changes in the morphology and cytoskeleton organisation of arterial endothelial cells. Eur J Vasc Endovasc Surg 9: 86‐92, 1995.
 53.Dal‐Bianco JP, Aikawa E, Bischoff J, Guerrero JL, Handschumacher MD, Sullivan S, Johnson B, Titus JS, Iwamoto Y, Wylie‐Sears J, Levine RA, Carpentier A. Active adaptation of the tethered mitral valve: Insights into a compensatory mechanism for functional mitral regurgitation. Circulation 120: 334‐342, 2009.
 54.David H, Boughner DR, Vesely I, Gerosa G. The pulmonary valve. Is it mechanically suitable for use as an aortic valve replacement? Asaio J 40: 206‐212, 1994.
 55.David Merryman W, Shadow Huang HY, Schoen FJ, Sacks MS. The effects of cellular contraction on aortic valve leaflet flexural stiffness. J Biomech 39: 88‐96, 2006.
 56.Davies PF. Mechanisms involved in endothelial responses to hemodynamic forces. Atherosclerosis 131(Suppl): S15‐17, 1997.
 57.Davies PF, Passerini AG, Simmons CA. Aortic valve: Turning over a new leaf(let) in endothelial phenotypic heterogeneity. Arterioscler Thromb Vasc Biol 24: 1331‐1333, 2004.
 58.Davies PF, Remuzzi A, Gordon EJ, Dewey CF, Jr, Gimbrone MA, Jr. Turbulent fluid shear stress induces vascular endothelial cell turnover in vitro. Proc Natl Acad Sci U S A 83: 2114‐2117, 1986.
 59.Davies PF, Shi C, Depaola N, Helmke BP, Polacek DC. Hemodynamics and the focal origin of atherosclerosis: A spatial approach to endothelial structure, gene expression, and function. Ann N Y Acad Sci 947: 7‐16; discussion 16‐17, 2001.
 60.Davies PF, Tripathi SC. Mechanical stress mechanisms and the cell. An endothelial paradigm. Circ Res 72: 239‐245, 1993.
 61.de la Pompa JL, Timmerman LA, Takimoto H, Yoshida H, Elia AJ, Samper E, Potter J, Wakeham A, Marengere L, Langille BL, Crabtree GR, Mak TW. Role of the NF‐ATc transcription factor in morphogenesis of cardiac valves and septum. Nature 392: 182‐186, 1998.
 62.de Lange FJ, Moorman AF, Anderson RH, Manner 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 95: 645‐654, 2004.
 63.Deck JD. Endothelial cell orientation on aortic valve leaflets. Cardiovasc Res 20: 760‐767, 1986.
 64.Dreger SA, Taylor PM, Allen SP, Yacoub MH. Profile and localization of matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) in human heart valves. J Heart Valve Dis 11: 875‐880; discussion 880, 2002.
 65.Driessen NJ, Boerboom RA, Huyghe JM, Bouten CV, Baaijens FP. Computational analyses of mechanically induced collagen fiber remodeling in the aortic heart valve. J Biomech Eng 125: 549‐557, 2003.
 66.Driessen NJ, Bouten CV, Baaijens FP. Improved prediction of the collagen fiber architecture in the aortic heart valve. J Biomech Eng 127: 329‐336, 2005.
 67.Einstein DR, Kunzelman KS, Reinhall PG, Cochran RP, Nicosia MA. Haemodynamic determinants of the mitral valve closure sound: A finite element study. Med Biol Eng Comput 42: 832‐846, 2004.
 68.Einstein DR, Kunzelman KS, Reinhall PG, Nicosia MA, Cochran RP. The relationship of normal and abnormal microstructural proliferation to the mitral valve closure sound. J Biomech Eng 127: 134‐147, 2005.
 69.Engelmayr GC, Jr., Hildebrand DK, Sutherland FW, Mayer JE, Jr., Sacks MS. A novel bioreactor for the dynamic flexural stimulation of tissue engineered heart valve biomaterials. Biomaterials 24: 2523‐2532, 2003.
 70.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 24: 527‐534, 2003.
 71.Fasol R, Meinhart J, Deutsch M, Binder T. Mitral valve repair with the Colvin‐Galloway Future Band. Ann Thorac Surg 77: 1985‐1988; discussion 1988, 2004.
 72.Filip DA, Radu A, Simionescu M. Interstitial cells of the heart valve possess characteristics similar to smooth muscle cells. Circ Res 59: 310‐320, 1986.
 73.Flameng W, Herijgers P, Bogaerts K. Recurrence of mitral valve regurgitation after mitral valve repair in degenerative valve disease. Circulation 107: 1609‐1613, 2003.
 74.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.
 75.Freed LA, Benjamin EJ, Levy D, Larson MG, Evans JC, Fuller DL, Lehman B, Levine RA. Mitral valve prolapse in the general population: The benign nature of echocardiographic features in the Framingham Heart Study. J Am Coll Cardiol 40: 1298‐1304, 2002.
 76.Freeman RV, Otto CM. Management of asymptomatic valvular aortic stenosis. Indian Heart J 54: 31‐38, 2002.
 77.Freeman RV, Otto CM. Spectrum of calcific aortic valve disease: Pathogenesis, disease progression, and treatment strategies. Circulation 111: 3316‐3326, 2005.
 78.Frisch‐Fay R. Flexible Bars. Washington, DC: Butterworths, 1962.
 79.Fung YC. What are the residual stresses doing in our blood vessels? Ann Biomed Eng 19: 237‐249, 1991.
 80.Fung YC, Liu SQ. Changes of zero‐stress state of rat pulmonary arteries in hypoxic hypertension. J Appl Physiol 70: 2455‐2470, 1991.
 81.Ge L, Sotiropoulos F. Direction and magnitude of blood flow shear stresses on the leaflets of aortic valves: Is there a link with valve calcification? J Biomech Eng 132: 014505, 2010.
 82.Gharacholou SM, Karon BL, Shub C, Pellikka PA. Aortic valve sclerosis and clinical outcomes: Moving toward a definition. Am J Med 124: 103‐110, 2011.
 83.Gillinov AM, Blackstone EH, White J, Howard M, Ahkrass R, Marullo A, Cosgrove DM. Durability of combined aortic and mitral valve repair. Ann Thorac Surg 72: 20‐27, 2001.
 84.Gillinov AM, Cosgrove DM, III, Shiota T, Qin J, Tsujino H, Stewart WJ, Thomas JD, Porqueddu M, White JA, Blackstone EH. Cosgrove‐Edwards Annuloplasty System: Midterm results. Ann Thorac Surg 69: 717‐721, 2000.
 85.Gillinov AM, Cosgrove DM, Blackstone EH, Diaz R, Arnold JH, Lytle BW, Smedira NG, Sabik JF, McCarthy PM, Loop FD. Durability of mitral valve repair for degenerative disease. J Thorac Cardiovasc Surg 116: 734‐743, 1998.
 86.Girard PR, Nerem RM. Shear stress modulates endothelial cell morphology and F‐actin organization through the regulation of focal adhesion‐associated proteins. J Cell Physiol 163: 179‐193, 1995.
 87.Gittenberger‐de Groot AC, Vrancken Peeters MP, Mentink MM, Gourdie RG, Poelmann RE. Epicardium‐derived cells contribute a novel population to the myocardial wall and the atrioventricular cushions. Circ Res 82: 1043‐1052, 1998.
 88.Gloeckner DC, Billiar KL, Sacks MS. Effects of mechanical fatigue on the bending properties of the porcine bioprosthetic heart valve. Asaio J 45: 59‐63, 1999.
 89.Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Huffman MD, Judd SE, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Mackey RH, Magid DJ, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER, III, Moy CS, Mussolino ME, Neumar RW, Nichol G, Pandey DK, Paynter NP, Reeves MJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Wong ND, Woo D, Turner MB. Heart disease and stroke statistics–2014 update: A report from the American Heart Association. Circulation 129: e28‐e292, 2014.
 90.Goldstein D, Moskowitz AJ, Gelijns AC, Ailawadi G, Parides MK, Perrault LP, Hung JW, Voisine P, Dagenais F, Gillinov AM, Thourani V, Argenziano M, Gammie JS, Mack M, Demers P, Atluri P, Rose EA, O'Sullivan K, Williams DL, Bagiella E, Michler RE, Weisel RD, Miller MA, Geller NL, Taddei‐Peters WC, Smith PK, Moquete E, Overbey JR, Kron IL, O'Gara PT, Acker MA. Two‐year outcomes of surgical treatment of severe ischemic mitral regurgitation. N Engl J Med 374: 344‐353, 2016.
 91.Gorman JH, III, Gupta KB, Streicher JT, Gorman RC, Jackson BM, Ratcliffe MB, Bogen DK, Edmunds LH, Jr. Dynamic three‐dimensional imaging of the mitral valve and left ventricle by rapid sonomicrometry array localization. J Thorac Cardiovasc Surg 112: 712‐726, 1996.
 92.Gorman JH, III, Jackson BM, Enomoto Y, Gorman RC. The effect of regional ischemia on mitral valve annular saddle shape. Ann Thorac Surg 77: 544‐548, 2004.
 93.Grande‐Allen KJ, Griffin BP, Ratliff NB, Cosgrove DM, Vesely I. Glycosaminoglycan profiles of myxomatous mitral leaflets and chordae parallel the severity of mechanical alterations. J Am Coll Cardiol 42: 271‐277, 2003.
 94.Grashow JS, Sacks MS, Liao J, Yoganathan AP. Planar biaxial creep and stress relaxation of the mitral valve anterior leaflet. Ann Biomed Eng 34: 1509‐1518, 2006a.
 95.Grashow JS, Yoganathan AP, Sacks MS. Biaxial stress‐stretch behavior of the mitral valve anterior leaflet at physiologic strain rates. Ann Biomed Eng 34: 315‐325, 2006b.
 96.Grau JB, Poggio P, Sainger R, Vernick WJ, Seefried WF, Branchetti E, Field BC, Bavaria JE, Acker MA, Ferrari G. Analysis of osteopontin levels for the identification of asymptomatic patients with calcific aortic valve disease. Ann Thorac Surg 93: 79‐86, 2012.
 97.Guilak F, Mow VC. The mechanical environment of the chondrocyte: A biphasic finite element model of cell‐matrix interactions in articular cartilage. J Biomech 33, 1663‐1673, 2000.
 98.Gupta V, Grande‐Allen KJ. Effects of static and cyclic loading in regulating extracellular matrix synthesis by cardiovascular cells. Cardiovasc Res 72: 375‐383, 2006.
 99.Gupta V, Werdenberg JA, Blevins TL, Grande‐Allen KJ. Synthesis of glycosaminoglycans in differently loaded regions of collagen gels seeded with valvular interstitial cells. Tissue Eng 13: 41‐49, 2007.
 100.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.
 101.Guy TS, Hill AC. Mitral valve prolapse. Annu Rev Med 63: 277‐292, 2012.
 102.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.
 103.Hartiala JJ, Mostbeck GH, Foster E, Fujita N, Dulce MC, Chazouilleres AF, Higgins CB. Velocity‐encoded cine MRI in the evaluation of left ventricular diastolic function: Measurement of mitral valve and pulmonary vein flow velocities and flow volume across the mitral valve. Am Heart J 125: 1054‐1066, 1993.
 104.Haussinger G, Pfennigs H, Chelapurath A, Reul H, Essers U. Influence of platelet aggregation inhibitors on platelet damage at prosthetic heart valves in‐vitro (author's transl). Biomed Tech (Berlin) 26: 99‐102, 1981.
 105.He Z, Ritchie J, Grashow JS, Sacks MS, Yoganathan AP. In vitro dynamic strain behavior of the mitral valve posterior leaflet. J Biomech Eng 127: 504‐511, 2005.
 106.He Z, Sacks MS, Baijens L, Wanant S, Shah P, Yoganathan AP. Effects of papillary muscle position on in‐vitro dynamic strain on the porcine mitral valve. J Heart Valve Dis 12: 488‐494, 2003.
 107.Helmlinger G, Geiger Rv, Fau‐Schreck S, Schreck S, Fau‐Nerem RM, Nerem RM. Effects of pulsatile flow on cultured vascular endothelial cell morphology. J Biomech Eng 113: 123‐131, 1991.
 108.Henney AM, Parker DJ, Davies MJ. Collagen biosynthesis in normal and abnormal human heart valves. Cardiovasc Res 16: 624‐630, 1982.
 109.Hilbert SL, Barrick, M.K., Ferrans, V.J. Porcine aortic valve bioprostheses: A morphologic comparison of the effects of fixation pressure. J Biomed Mater Res 24: 773‐787, 1990.
 110.Hilbert SL, Ferrans VJ, Swanson WM. Optical methods for the nondestructive evaluation of collagen morphology in bioprosthetic heart valves. J Biomed Mater Res 20: 1411‐1421, 1986.
 111.Hinton RB, Jr., Lincoln J, Deutsch GH, Osinska H, Manning PB, Benson DW, Yutzey KE. Extracellular matrix remodeling and organization in developing and diseased aortic valves. Circ Res 98: 1431‐1438, 2006.
 112.Hinton RB, Yutzey KE. Heart valve structure and function in development and disease. Annu Rev Physiol 73: 29‐46, 2011.
 113.Hoffman JI, Kaplan S. The incidence of congenital heart disease. J Am Coll Cardiol 39: 1890‐1900, 2002.
 114.Huang HY, Liao J, Sacks MS. In-situ deformation of the aortic valve interstitial cell nucleus under diastolic loading. J Biomech Eng 129: 880‐889, 2007.
 115.Hurle JM, Kitten GT, Sakai LY, Volpin D, Solursh M. Elastic extracellular matrix of the embryonic chick heart: An immunohistological study using laser confocal microscopy. Dev Dyn 200: 321‐332, 1994.
 116.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.
 117.Jiang X, Rowitch DH, Soriano P, McMahon AP, Sucov HM. Fate of the mammalian cardiac neural crest. Development 127: 1607‐1616, 2000.
 118.Johnson CM, Fass DN. Porcine cardiac valvular endothelial cells in culture. A relative deficiency of fibronectin synthesis in vitro. Lab Invest 49: 589‐598, 1983.
 119.Joyce EM, Liao J, Schoen FJ, Mayer JE, Jr., Sacks MS. Functional collagen fiber architecture of the pulmonary heart valve cusp. Ann Thorac Surg 87: 1240‐1249, 2009.
 120.Juliano RL, Haskill S. Signal transduction from the extracellular matrix. J Cell Biol 120: 577‐585, 1993.
 121.Kilner PJ, Yang GZ, Mohiaddin RH, Firmin DN, Longmore DB. Helical and retrograde secondary flow patterns in the aortic arch studied by three‐directional magnetic resonance velocity mapping. Circulation 88: 2235‐2247, 1993.
 122.Kim WY, Walker PG, Pedersen EM, Poulsen JK, Oyre S, Houlind K, Yoganathan AP. Left ventricular blood flow patterns in normal subjects: A quantitative analysis by three‐dimensional magnetic resonance velocity mapping. J Am Coll Cardiol 26: 224‐238, 1995.
 123.Konduri S, Xing Y, Warnock JN, He Z, Yoganathan AP. Normal physiological conditions maintain the biological characteristics of porcine aortic heart valves: An ex vivo organ culture study. Ann Biomed Eng 33: 1158‐1166, 2005.
 124.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 71: 548‐556, 2006.
 125.Kunzelman KS, Cochran RP, Chuong C, Ring WS, Verrier ED, Eberhart RD. Finite element analysis of the mitral valve. J Heart Valve Dis 2: 326‐340, 1993.
 126.Kunzelman KS, Cochran RP, Verrier ED, Eberhart RC. Anatomic basis for mitral valve modelling. J Heart Valve Dis 3: 491‐496, 1994.
 127.Kunzelman KS, Einstein DR, Cochran RP. Fluid‐structure interaction models of the mitral valve: Function in normal and pathological states. Philos Trans R Soc Lond B Biol Sci 362: 1393‐1406, 2007.
 128.Kunzelman KS, Quick DW, Cochran RP. Altered collagen concentration in mitral valve leaflets: Biochemical and finite element analysis. Ann Thorac Surg 66: S198‐S205, 1998.
 129.Kurtz CE, Otto CM. Aortic stenosis: Clinical aspects of diagnosis and management, with 10 illustrative case reports from a 25‐year experience. Medicine 89: 349‐379, 2010.
 130.Laforest B, Andelfinger G, Nemer M. Loss of Gata5 in mice leads to bicuspid aortic valve. J Clin Invest 121: 2876‐2887, 2011.
 131.Lanir Y. Mechanisms of residual stress in soft tissues. J Biomech Eng 131: 044506, 2009.
 132.Leask RL, Jain N, Butany J. Endothelium and valvular diseases of the heart. Microsc Res Tech 60: 129‐137, 2003.
 133.Lee CH, Amini R, Gorman RC, Gorman JH, III, Sacks MS. An inverse modeling approach for stress estimation in mitral valve anterior leaflet valvuloplasty for in‐vivo valvular biomaterial assessment. J Biomech 47: 2055‐2063, 2014.
 134.Lee CH, Carruthers CA, Ayoub S, Gorman RC, Gorman JH, III, Sacks MS. Quantification and simulation of layer‐specific mitral valve interstitial cells deformation under physiological loading. J Theor Biol 373: 26‐39, 2015.
 135.Lee CH, Zhang W, Liao J, Carruthers CA, Sacks JI, Sacks MS. On the presence of affine fibril and fiber kinematics in the mitral valve anterior leaflet. Biophys J 108: 2074‐2087, 2015.
 136.Lee TC, Zhao YD, Courtman DW, Stewart DJ. Abnormal aortic valve development in mice lacking endothelial nitric oxide synthase. Circulation 101: 2345‐2348, 2000.
 137.Lehoux S, Tedgui A. Cellular mechanics and gene expression in blood vessels. J Biomech 36: 631‐643, 2003.
 138.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 103: 948‐956, 2008.
 139.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, Marec HL, Tourneau TL, 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.
 140.Li C, Xu Q. Mechanical stress‐initiated signal transductions in vascular smooth muscle cells. Cell Signal 12: 435‐445, 2000.
 141.Liao J, Yang L, Grashow J, Sacks MS. The relation between collagen fibril kinematics and mechanical properties in the mitral valve anterior leaflet. J Biomech Eng 129: 78‐87, 2007.
 142.Liebner S, Cattelino A, Gallini R, Rudini N, Iurlaro M, Piccolo S, Dejana E. Beta‐catenin is required for endothelial‐mesenchymal transformation during heart cushion development in the mouse. J Cell Biol 166: 359‐367, 2004.
 143.Lincoln J, Alfieri CM, Yutzey KE. BMP and FGF regulatory pathways control cell lineage diversification of heart valve precursor cells. Dev Biol 292: 292‐302, 2006.
 144.Lincoln J, Alfieri CM, Yutzey KE. Development of heart valve leaflets and supporting apparatus in chicken and mouse embryos. Dev Dyn 230: 239‐250, 2004.
 145.Lincoln J, Kist R, Scherer G, Yutzey KE. Sox9 is required for precursor cell expansion and extracellular matrix organization during mouse heart valve development. Dev Biol 305: 120‐132, 2007.
 146.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.
 147.Liu SQ, Fung YC. Relationship between hypertension, hypertrophy, and opening angle of zero‐stress state of arteries following aortic constriction. J Biomech Eng 111: 325‐335, 1989.
 148.Lloyd‐Jones D, Adams RJ, Brown TM, Carnethon M, Dai S, De Simone G, Ferguson TB, Ford E, Furie K, Gillespie C, Go A, Greenlund K, Haase N, Hailpern S, Ho PM, Howard V, Kissela B, Kittner S, Lackland D, Lisabeth L, Marelli A, McDermott MM, Meigs J, Mozaffarian D, Mussolino M, Nichol G, Roger VL, Rosamond W, Sacco R, Sorlie P, Stafford R, Thom T, Wasserthiel‐Smoller S, Wong ND, Wylie‐Rosett J. Executive summary: Heart disease and stroke statistics–2010 update: A report from the American Heart Association. Circulation 121: 948‐954, 2010.
 149.Lo D, Vesely I. Biaxial strain analysis of the porcine aortic valve. Ann Thorac Surg 60: S374‐S378, 1995.
 150.Loukas M, Bilinsky E, Bilinsky S, Blaak C, Tubbs RS, Anderson RH. The anatomy of the aortic root. Clin Anat 27: 748‐756, 2014.
 151.Luscinskas FW, Lawler J. Integrins as dynamic regulators of vascular function. FASEB J 8: 929‐938, 1994.
 152.Ma L, Lu M‐F, Schwartz RJ, Martin JF. Bmp2 is essential for cardiac cushion epithelial‐mesenchymal transition and myocardial patterning. Development 132: 5601‐5611, 2005.
 153.Mahimkar R, Nguyen A, Mann M, Yeh CC, Zhu BQ, Karliner JS, Lovett DH. Cardiac transgenic matrix metalloproteinase‐2 expression induces myxomatous valve degeneration: A potential model of mitral valve prolapse disease. Cardiovasc Pathol 18: 253‐261, 2009.
 154.Maisano F, Redaelli A, Soncini M, Votta E, Arcobasso L, Alfieri O. An annular prosthesis for the treatment of functional mitral regurgitation: Finite element model analysis of a dog bone‐shaped ring prosthesis. Ann Thorac Surg 79: 1268‐1275, 2005.
 155.Mansi T, Voigt I, Georgescu B, Zheng X, Mengue EA, Hackl M, Ionasec RI, Noack T, Seeburger J, Comaniciu D. An integrated framework for finite‐element modeling of mitral valve biomechanics from medical images: Application to MitralClip intervention planning. Med Image Anal 16: 1330‐1346, 2012.
 156.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.
 157.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 94: 368‐375, 1996.
 158.May‐Newman K, Yin FC. Biaxial mechanical behavior of excised porcine mitral valve leaflets. Am J Physiol 269: H1319‐H1327, 1995.
 159.May‐Newman K, Yin FC. A constitutive law for mitral valve tissue. J Biomech Eng 120: 38‐47, 1998.
 160.Mayne AS, Christie GW, Smaill BH, Hunter PJ, Barratt‐Boyes BG. An assessment of the mechanical properties of leaflets from four second‐generation porcine bioprostheses with biaxial testing techniques. J Thorac Cardiovasc Surg 98: 170‐180, 1989.
 161.McGee EC, Gillinov AM, Blackstone EH, Rajeswaran J, Cohen G, Najam F, Shiota T, Sabik JF, Lytle BW, McCarthy PM, Cosgrove DM. Recurrent mitral regurgitation after annuloplasty for functional ischemic mitral regurgitation. J Thorac Cardiovasc Surg 128: 916‐924, 2004.
 162.Merryman WD, Huang H‐YS, Schoen FJ, Sacks MS. The effects of cellular contraction on aortic valve leaflet flexural stiffness. J Biomech 39: 88‐96, 2006.
 163.Merryman WD, Liao J, Parekh A, Candiello JE, Lin H, Sacks MS. Differences in tissue‐remodeling potential of aortic and pulmonary heart valve interstitial cells. Tissue Eng 13: 2281‐2289, 2007.
 164.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.
 165.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 290: H224‐H231, 2006.
 166.Messier RH, Jr., Bass BL, Aly HM, Jones JL, Domkowski PW, Wallace RB, Hopkins RA. Dual structural and functional phenotypes of the porcine aortic valve interstitial population: Characteristics of the leaflet myofibroblast. J Surg Res 57: 1‐21, 1994.
 167.Ming L, Zhen HK. Study of the closing mechanism of natural heart valves. Appl Math Mec 7, 955‐964, 1986.
 168.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.
 169.Montero JA, Giron B, Arrechedera H, Cheng YC, Scotting P, Chimal‐Monroy J, Garcia‐Porrero JA, Hurle JM. Expression of Sox8, Sox9 and Sox10 in the developing valves and autonomic nerves of the embryonic heart. Mech Dev 118: 199‐202, 2002.
 170.Moura LM, Ramos SF, Zamorano JL, Barros IM, Azevedo LF, Rocha‐Goncalves F, Rajamannan NM. Rosuvastatin affecting aortic valve endothelium to slow the progression of aortic stenosis. J Am Coll Cardiol 49: 554‐561, 2007.
 171.Mulholland DL, Gotlieb AI. Cell biology of valvular interstitial cells. Can J Cardiol 12: 231‐236, 1996.
 172.Nakamura T, Colbert MC, Robbins J. Neural crest cells retain multipotential characteristics in the developing valves and label the cardiac conduction system. Circ Res 98: 1547‐1554, 2006.
 173.Nicosia MA, Cochran RP, Einstein DR, Rutland CJ, Kunzelman KS. A coupled fluid‐structure finite element model of the aortic valve and root. J Heart Valve Dis 12: 781‐789, 2003.
 174.Niederer SA, Smith NP. At the heart of computational modelling. J Physiol 590: 1331‐1338, 2012.
 175.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.
 176.Nus M, MacGrogan D, Martinez‐Poveda B, Benito Y, Casanova JC, Fernandez‐Aviles F, Bermejo J, de la Pompa JL. Diet‐induced aortic valve disease in mice haploinsufficient for the Notch pathway effector RBPJK/CSL. Arterioscler Thromb Vasc Biol 31: 1580‐1588, 2011.
 177.Ogden R. Nonlinear elasticity, anisotropy, material stability, and residual stresses in soft tissue. In: Ogden RW, editor. Biomechanics of Soft Tissue in Cardiovascular System. New York: Springer, 2003.
 178.Oh JK, Appleton CP, Hatle LK, Nishimura RA, Seward JB, Tajik AJ. The noninvasive assessment of left ventricular diastolic function with two‐dimensional and Doppler echocardiography. J Am Soc Echocardiogr 10: 246‐270, 1997.
 179.Ormiston JA, Shah PM, Tei C, Wong M. Size and motion of the mitral valve annulus in man. I. A two‐dimensional echocardiographic method and findings in normal subjects. Circulation 64: 113‐120, 1981.
 180.Otsuji Y, Handschumacher MD, Schwammenthal E, Jiang L, Song JK, Guerrero JL, Vlahakes GJ, Levine RA. Insights from three‐dimensional echocardiography into the mechanism of functional mitral regurgitation: Direct in vivo demonstration of altered leaflet tethering geometry. Circulation 96: 1999‐2008, 1997.
 181.Otto CM. Clinical practice. Evaluation and management of chronic mitral regurgitation. N Engl J Med 345: 740‐746, 2001.
 182.Otto CM. Calcific aortic valve disease: Outflow obstruction is the end stage of a systemic disease process. Eur Heart J 30: 1940‐1942, 2009.
 183.Otto CM. Calcific aortic valve disease: New concepts. Semin Thorac Cardiovasc Surg 22: 276‐284, 2010.
 184.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.
 185.Owens DS, Otto CM. Is it time for a new paradigm in calcific aortic valve disease? JACC Cardiovasc Imaging 2: 928‐930, 2009.
 186.Parolari A, Tremoli E, Cavallotti L, Trezzi M, Kassem S, Loardi C, Veglia F, Ferrari G, Pacini D, Alamanni F. Do statins improve outcomes and delay the progression of non‐rheumatic calcific aortic stenosis? Heart (British Cardiac Society) 97: 523‐529, 2011.
 187.Perez‐Pomares JM, Gonzalez‐Rosa JM, Munoz‐Chapuli R. Building the vertebrate heart ‐ an evolutionary approach to cardiac development. Int J Dev Biol 53: 1427‐1443, 2009.
 188.Person AD, Klewer SE, Runyan RB. Cell biology of cardiac cushion development. Int Rev Cytol 243: 287‐335, 2005.
 189.Pflederer T, Achenbach S. Aortic valve stenosis: CT contributions to diagnosis and therapy. J Cardiovasc Comput Tomogr 4: 355‐364, 2010.
 190.Pho M, Lee W, Watt DR, Laschinger C, Simmons CA, McCulloch CA. Cofilin is a marker of myofibroblast differentiation in cells from porcine aortic cardiac valves. Am J Physiol Heart Circ Physiol 294: H1767‐H1778, 2008.
 191.Pierlot CM, Lee JM, Amini R, Sacks MS, Wells SM. Pregnancy‐induced remodeling of collagen architecture and content in the mitral valve. Ann Biomed Eng 42: 2058‐2071, 2014.
 192.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.
 193.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.
 194.Prasad AR, Logan SA, Nerem RM, Schwartz CJ, Sprague EA. Flow‐related responses of intracellular inositol phosphate levels in cultured aortic endothelial cells. Circ Res 72: 827‐836, 1993.
 195.Prot V, Haaverstad R, Skallerud B. Finite element analysis of the mitral apparatus: Annulus shape effect and chordal force distribution. Biomech Model Mechanobiol 8: 43‐55, 2009.
 196.Prot V, Skallerud B. Nonlinear solid finite element analysis of mitral valves with heterogeneous leaflet layers. Comput Mech 43: 353‐368, 2009.
 197.Prot V, Skallerud B, Holzapfel G. Transversely isotropic membrane shells with application to mitral valve mechanics. Constitutive modelling and finite element implementation. Int J Numer Methods Eng 71: 987‐1008, 2007.
 198.Pye MP, Pringle SD, Cobbe SM. Reference values and reproducibility of Doppler echocardiography in the assessment of the tricuspid valve and right ventricular diastolic function in normal subjects. Am J Cardiol 67: 269‐273, 1991.
 199.Quick DW, Kunzelman KS, Kneebone JM, Cochran RP. Collagen synthesis is upregulated in mitral valves subjected to altered stress. Asaio J 43: 181‐186, 1997.
 200.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.
 201.Rabkin E, Hoerstrup SP, Aikawa M, Mayer JE, Jr., Schoen FJ. Evolution of cell phenotype and extracellular matrix in tissue‐engineered heart valves during in‐vitro maturation and in‐vivo remodeling. J Heart Valve Dis 11: 308‐314; discussion 314, 2002.
 202.Rabkin‐Aikawa E, Aikawa M, Farber M, Kratz JR, Garcia‐Cardena G, Kouchoukos NT, Mitchell MB, Jonas RA, Schoen FJ. Clinical pulmonary autograft valves: Pathologic evidence of adaptive remodeling in the aortic site. J Thorac Cardiovasc Surg 128: 552‐561, 2004.
 203.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 13: 841‐847, 2004.
 204.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.
 205.Rajamannan NM, Subramaniam M, Rickard D, Stock SR, Donovan J, Springett M, Orszulak T, Fullerton DA, Tajik AJ, Bonow RO, Spelsberg T. Human aortic valve calcification is associated with an osteoblast phenotype. Circulation 107: 2181‐2184, 2003.
 206.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 392: 186‐190, 1998.
 207.Rausch MK, Famaey N, Shultz TO, Bothe W, Miller DC, Kuhl E. Mechanics of the mitral valve: A critical review, an in vivo parameter identification, and the effect of prestrain. Biomech Model Mechanobiol 12: 1053‐1071, 2012.
 208.Rausch MK, Tibayan FA, Miller DC, Kuhl E. Evidence of adaptive mitral leaflet growth. J Mech Behav Biomed Mater 15: 208‐217, 2012.
 209.Reul H, Talukder N. Heart valve mechanics. In: The Heart (7th ed). McGraw Hill, 1989.
 210.Roberts WC, Ko JM. Frequency by decades of unicuspid, bicuspid, and tricuspid aortic valves in adults having isolated aortic valve replacement for aortic stenosis, with or without associated aortic regurgitation. Circulation 111: 920‐925, 2005.
 211.Rodriguez KJ, Piechura LM, Masters KS. Regulation of valvular interstitial cell phenotype and function by hyaluronic acid in 2‐D and 3‐D culture environments. Matrix Biol 30: 70‐82, 2011.
 212.Rossebo AB, Pedersen TR, Boman K, Brudi P, Chambers JB, Egstrup K, Gerdts E, Gohlke‐Barwolf C, Holme I, Kesaniemi YA, Malbecq W, Nienaber CA, Ray S, Skjaerpe T, Wachtell K, Willenheimer R. Intensive lipid lowering with simvastatin and ezetimibe in aortic stenosis. N Engl J Med 359: 1343‐1356, 2008.
 213.Ruwhof C, van der Laarse A. Mechanical stress‐induced cardiac hypertrophy: Mechanisms and signal transduction pathways. Cardiovasc Res 47: 23‐37, 2000.
 214.Sacks MS, Enomoto Y, Graybill JR, Merryman WD, Zeeshan A, Yoganathan AP, Levy RJ, Gorman RC, Gorman JH, III. In‐vivo dynamic deformation of the mitral valve anterior leaflet. Ann Thorac Surg 82: 1369‐1377, 2006.
 215.Sacks MS, He Z, Baijens L, Wanant S, Shah P, Sugimoto H, Yoganathan AP. Surface strains in the anterior leaflet of the functioning mitral valve. Ann Biomed Eng 30: 1281‐1290, 2002.
 216.Sacks MS, Merryman WD, Schmidt DE. On the biomechanics of heart valve function. J Biomech 42: 1804‐1824, 2009.
 217.Sacks MS, Smith DB, Hiester ED. The aortic valve microstructure: Effects of transvalvular pressure. J Biomed Mater Res 41: 131‐141, 1998.
 218.Sacks MS, Yoganathan AP. Heart valve function: A biomechanical perspective. Philos Trans R Soc Lond B Biol Sci 363: 2481, 2008.
 219.Sadoshima J, Izumo S. The cellular and molecular response of cardiac myocytes to mechanical stress. Annu Rev Physiol 59: 551‐571, 1997.
 220.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.
 221.Sakamoto Y, Buchanan RM, Sacks MS. On intrinsic stress fiber contractile forces in semilunar heart valve interstitial cells using a continuum mixture model. J Mech Behav Biomed Mater 54: 244‐258, 2016.
 222.Schmidt C, Pommerenke H, Durr F, Nebe B, Rychly J. Mechanical stressing of integrin receptors induces enhanced tyrosine phosphorylation of cytoskeletally anchored proteins. J Biol Chem 273: 5081‐5085, 1998.
 223.Schmidtke C, Poppe D, Dahmen G, Sievers HH. Echocardiographic and hemodynamic characteristics of reconstructed bicuspid aortic valves at rest and exercise. Z Kardiol 94: 437‐444, 2005.
 224.Schoen F. Aortic valve structure‐function correlations: Role of elastic fibers no longer a stretch of the imagination. J Heart Valve Dis 6: 1‐6, 1997.
 225.Schoen FJ. Cardiac valves and valvular pathology: Update on function, disease, repair, and replacement. Cardiovasc Pathol 14: 189‐194, 2005.
 226.Schoen FJ. Evolving concepts of cardiac valve dynamics: The continuum of development, functional structure, pathobiology, and tissue engineering. Circulation 118: 1864‐1880, 2008.
 227.Schwammenthal E, Chen C, Benning F, Block M, Breithardt G, Levine RA. Dynamics of mitral regurgitant flow and orifice area. Physiologic application of the proximal flow convergence method: Clinical data and experimental testing. Circulation 90: 307‐322, 1994.
 228.Shelton EL, Yutzey KE. Twist1 function in endocardial cushion cell proliferation, migration, and differentiation during heart valve development. Dev Biol 317: 282‐295, 2008.
 229.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 96: 792‐799, 2005.
 230.Skallerud B, Prot V, Nordrum IS. Modeling active muscle contraction in mitral valve leaflets during systole: A first approach. Biomech Model Mechanobiol 10: 11‐26, 2011.
 231.Sloth E, Houlind KC, Oyre S, Kim WY, Pedersen EM, Jorgensen HS, Hasenkam JM. Three‐dimensional visualization of velocity profiles in the human main pulmonary artery with magnetic resonance phase‐velocity mapping. Am Heart J 128: 1130‐1138, 1994.
 232.Smith DB, Sacks MS, Vorp DA, Thornton M. Surface geometric analysis of anatomic structures using biquintic finite element interpolation. Ann Biomed Eng 28: 598‐611, 2000.
 233.Snarr BS, Kern CB, Wessels A. Origin and fate of cardiac mesenchyme. Dev Dyn 237: 2804‐2819, 2008.
 234.Stamatas GN, McIntire LV. Rapid flow‐induced responses in endothelial cells. Biotechnol Prog 17: 383‐402, 2001.
 235.Stella JA, Sacks MS. On the biaxial mechanical properties of the layers of the aortic valve leaflet. J Biomech Eng 129: 757‐766, 2007.
 236.Stephens E, Durst C, Swanson J, Grande‐Allen KJ, Ingels N, Jr., Miller DC. Functional coupling of valvular interstitial cells and collagen via α2β1 integrins in the mitral leaflet. Cel Mol Bioeng 3: 428‐437, 2010.
 237.Stephens E, Grande‐Allen K. Age‐related changes in collagen synthesis and turnover in porcine heart valves. J Heart Valve Dis 16: 672‐682, 2007.
 238.Stephens EH, Chu CK, Grande‐Allen KJ. Valve proteoglycan content and glycosaminoglycan fine structure are unique to microstructure, mechanical loads, and age: Relevance to an age‐specific tissue engineered heart valve. Acta Biomaterial 4: 1148‐1160, 2008.
 239.Stephens EH, de Jonge N, McNeill MP, Durst CA, Grande‐Allen KJ. Age‐related changes in material behavior of porcine mitral and aortic valves and correlation to matrix composition. Tissue Eng Part A 16: 867‐878, 2010.
 240.Stephens EH, Durst CA, West JL, Grande‐Allen KJ. Mitral valvular interstitial cell responses to substrate stiffness depend on age and anatomic region. Acta Biomater 7: 75‐82, 2011.
 241.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.
 242.Stephens EH, Saltarrelli JG, Baggett LS, Nandi I, Kuo JJ, Davis AR, Olmsted‐Davis EA, Reardon MJ, Morrisett JD, Grande‐Allen KJ. Differential proteoglycan and hyaluronan distribution in calcified aortic valves. Cardiovasc Pathol 20: 334‐342, 2011.
 243.Stephens EH, Timek TA, Daughters GT, Kuo JJ, Patton AM, Baggett LS, Ingels NB, Miller DC, Grande‐Allen KJ. Significant changes in mitral valve leaflet matrix composition and turnover with tachycardia‐induced cardiomyopathy. Circulation 120: S112‐S119, 2009.
 244.Stevanella M, Maffessanti F, Conti CA, Votta E, Arnoldi A, Lombardi M, Parodi O, Caiani EG, Redaelli A. Mitral valve patient‐specific finite element modeling from cardiac MRI: Application to an annuloplasty procedure. Cardiovas Eng Tech 2: 66‐76, 2011.
 245.Stewart BF, Siscovick D, Lind BK, Gardin JM, Gottdiener JS, Smith VE, Kitzman DW, Otto CM. Clinical factors associated with calcific aortic valve disease. Cardiovascular Health Study. J Am Coll Cardiol 29: 630‐634, 1997.
 246.Stradins P, Lacis R, Ozolanta I, Purina B, Ose V, Feldmane L, Kasyanov V. Comparison of biomechanical and structural properties between human aortic and pulmonary valve. Eur J Cardiothorac Surg 26: 634‐639, 2004.
 247.Sun W, Martin C, Pham T. Computational modeling of cardiac valve function and intervention. Annu Rev Biomed Eng 16: 53‐76, 2014.
 248.Sung HW, Philpot EF, Nanda NC, Yoganathan AP. Axial flow velocity patterns in a pulmonary artery model with varying degrees of valvular pulmonic stenosis: Pulsatile in vitro studies. J Biomech 23: 563‐578, 1990.
 249.Sung HW, Yoganathan AP. Axial flow velocity patterns in a normal human pulmonary artery model: Pulsatile in vitro studies. J Biomech 23: 201‐214, 1990.
 250.Sung HW, Yoganathan AP. Secondary flow velocity patterns in a pulmonary artery model with varying degrees of valvular pulmonic stenosis: Pulsatile in vitro studies. J Biomech Eng 112: 88‐92, 1990.
 251.Taber LA, Humphrey JD. Stress‐modulated growth, residual stress, and vascular heterogeneity. J Biomech Eng 123: 528‐535, 2001.
 252.Tanaka K, Sata M, Fukuda D, Suematsu Y, Motomura N, Takamoto S, Hirata Y, Nagai R. Age‐associated aortic stenosis in apolipoprotein E‐deficient mice. J Am Coll Cardiol 46: 134‐141, 2005.
 253.Tao G, Kotick JD, Lincoln J. Heart valve development, maintenance, and disease: The role of endothelial cells. Curr Top Dev Biol 100: 203‐232, 2012.
 254.Taylor PM, Batten P, Brand NJ, Thomas PS, Yacoub MH. The cardiac valve interstitial cell. Int J Biochem Cell Biol 35: 113‐118, 2003.
 255.Thubrikar M, Aouad J, Nolan SP. Comparison of the in‐vivo and in‐vitro mechanical properties of aortic valve leafelts. J Thorac Cardiovasc Surg 92, 1986.
 256.Thubrikar M, Bosher LP, Harry RR, Nolan SP. Mechanism of opening of the natural aortic valve in relation to the design of trileaflet prostheses. Surg Forum 28: 264‐266, 1977.
 257.Thubrikar M, Bosher LP, Nolan SP. The mechanism of opening of the aortic valve. J Thorac Cardiovasc Surg 77: 863‐870, 1979.
 258.Thubrikar M, Carabello BA, Aouad J, Nolan SP. Interpretation of aortic root angiography in dogs and in humans. Cardiovasc Res 16: 16‐21, 1982.
 259.Thubrikar M, Harry R, Nolan SP. Normal aortic valve function in dogs. Am J Cardiol 40: 563‐568, 1977.
 260.Thubrikar M, Nolan SP, Bosher LP, Deck JD. The cyclic changes and structure of the base of the aortic valve. Am Heart J 99: 217‐224, 1980.
 261.Thubrikar M, Piepgrass W, Bosher L, Nolan S. The elastic modulus of canine aortic valve leaflets in vivo and in vitro. Circ Res 47: 792‐800, 1980.
 262.Thubrikar M, Piepgrass W, Deck J, Nolan S. Stresses of natural versus prosthetic aortic valve leaflets in vivo. Ann Thorac Surg 30: 230‐239, 1980.
 263.Thubrikar M, Piepgrass WC, Shaner TW, Nolan SP. Design and dynamic variations of aortic valve leaflets in vivo. Surg Forum 30: 241‐243, 1979.
 264.Thubrikar M, Piepgrass WC, Shaner TW, Nolan SP. The design of the normal aortic valve. Am J Physiol 241: H795‐H801, 1981.
 265.Thubrikar M, Skinner J, Eppink R, Nolan S. Stress analysis of porcine bioprosthetic heart valves in vivo. J Biomed Mater Res 16: 811, 1982.
 266.Thubrikar M, Skinner JR, Aouad J, Finkelmeier BA, Nolan SP. Analysis of the design and dynamics of aortic bioprostheses in vivo. J Thorac Cardiovasc Surg 84: 282‐290, 1982.
 267.Thubrikar MJ, Aouad J, Nolan SP. Comparison of the in vivo and in vitro mechanical properties of aortic valve leaflets. J Thorac Cardiovasc Surg 92: 29‐36, 1986.
 268.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 58: 304‐308, 1986.
 269.Thubrikar MJ, Nolan SP, Aouad J, Deck JD. Stress sharing between the sinus and leaflets of canine aortic valve. Ann Thorac Surg 42: 434‐440, 1986.
 270.Tibayan FA, Rodriguez F, Langer F, Zasio MK, Bailey L, Liang D, Daughters GT, Ingels NB, Jr., Miller DC. Annular remodeling in chronic ischemic mitral regurgitation: Ring selection implications. Ann Thorac Surg 76: 1549‐1554; discussion 1554‐1545, 2003.
 271.Timek TA, Dagum P, Lai DT, Liang D, Daughters GT, Tibayan F, Ingels NB, Jr., Miller DC. Tachycardia‐induced cardiomyopathy in the ovine heart: Mitral annular dynamic three‐dimensional geometry. J Thorac Cardiovasc Surg 125: 315‐324, 2003.
 272.Timek TA, Lai DT, Dagum P, Liang D, Daughters GT, Ingels NB, Jr., Miller DC. Mitral leaflet remodeling in dilated cardiomyopathy. Circulation 114: I518‐I523, 2006.
 273.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.
 274.Towler DA. Molecular and cellular aspects of calcific aortic valve disease. Circ Res 113: 198‐208, 2013.
 275.Umesan CV, Kapoor A, Sinha N, Kumar AS, Goel PK. Effect of Inoue balloon mitral valvotomy on severe pulmonary arterial hypertension in 315 patients with rheumatic mitral stenosis: Immediate and long‐term results. J Heart Valve Dis 9: 609‐615, 2000.
 276.Verzi MP, McCulley DJ, De Val S, Dodou E, Black BL. The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field. Dev Biol 287: 134‐145, 2005.
 277.Vesely I. The role of elastin in aortic valve mechanics. J Biomech 31: 115‐123, 1998.
 278.Vesely I, Casarotto DC, Gerosa G. Mechanics of cryopreserved aortic and pulmonary homografts. J Heart Valve Dis 9: 27‐37, 2000.
 279.Vesely I, Mako WJ. Comparison of the compressive buckling of porcine aortic valve cusps and bovine pericardium. J Heart Valve Dis 7: 34‐39, 1998.
 280.Vesely I, Noseworthy R. Micromechanics of the fibrosa and the ventricularis in aortic valve leaflets. J Biomech 25: 101‐113, 1992.
 281.Votta E, Caiani E, Veronesi F, Soncini M, Montevecchi FM, Redaelli A. Mitral valve finite‐element modelling from ultrasound data: A pilot study for a new approach to understand mitral function and clinical scenarios. Philos Trans A Math Phys Eng Sci 366: 3411‐3434, 2008.
 282.Votta E, Le TB, Stevanella M, Fusini L, Caiani EG, Redaelli A, Sotiropoulos F. Toward patient‐specific simulations of cardiac valves: State‐of‐the‐art and future directions. J Biomech 46: 217‐228, 2013.
 283.Votta E, Maisano F, Bolling SF, Alfieri O, Montevecchi FM, Redaelli A. The Geoform disease‐specific annuloplasty system: A finite element study. Ann Thorac Surg 84: 92‐101, 2007.
 284.Wang Q, Sun W. Finite element modeling of mitral valve dynamic deformation using patient‐specific multi‐slices computed tomography scans. Ann Biomed Eng 41: 142‐153, 2013.
 285.Weinberg EJ, Mofrad MRK. Transient, three‐dimensional, multiscale simulations of the human aortic valve. Cardiovasc Eng 7: 140‐155, 2007.
 286.Weinberg EJ, Shahmirzadi D, Mofrad MRK. On the multiscale modeling of heart valve biomechanics in health and disease. Biomech Model Mechanobiol 9: 373‐387, 2010.
 287.Weiss RM, Ohashi M, Miller JD, Young SG, Heistad DD. Calcific aortic valve stenosis in old hypercholesterolemic mice. Circulation 114: 2065‐2069, 2006.
 288.Wells SM, Pierlot CM, Moeller AD. Physiological remodeling of the mitral valve during pregnancy. Am J Physiol Heart Circ Physiol 303: H878‐H892, 2012.
 289.Wenk JF, Zhang Z, Cheng G, Malhotra D, Acevedo‐Bolton G, Burger M, Suzuki T, Saloner DA, Wallace AW, Guccione JM, Ratcliffe MB. First finite element model of the left ventricle with mitral valve: Insights into ischemic mitral regurgitation. Ann Thorac Surg 89: 1546‐1553, 2010.
 290.Weyman AE. Principles and Practices of Echocardiography. Philadelphia, PA: Lea & Febiger, 1994.
 291.Willems IE, Havenith MG, Smits JF, Daemen MJ. Structural alterations in heart valves during left ventricular pressure overload in the rat. Lab Invest 71: 127‐133, 1994.
 292.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.
 293.Wyss K, Yip CY, Mirzaei Z, Jin X, Chen J‐H, Simmons CA. The elastic properties of valve interstitial cells undergoing pathological differentiation. J Biomech 45: 882‐887, 2012.
 294.Xie GY, Bhakta D, Smith MD. Echocardiographic follow‐up study of the Ross procedure in older versus younger patients. Am Heart J 142: 331‐335, 2001.
 295.Yacoub MH, Cohn LH. Novel approaches to cardiac valve repair: From structure to function: Part II. Circulation 109: 1064‐1072, 2004.
 296.Yang J, Weinberg RA. Epithelial‐mesenchymal transition: At the crossroads of development and tumor metastasis. Dev Cell 14: 818‐829, 2008.
 297.Yellin EL, Peskin C, Yoran C, Koenigsberg M, Matsumoto M, Laniado S, McQueen D, Shore D, Frater RW. Mechanisms of mitral valve motion during diastole. Am J Physiol 241: H389‐H400, 1981.
 298.Yoganathan AP. Fluid mechanics of aortic stenosis. Eur Heart J 9(Suppl E): 13‐17, 1988.
 299.Zhang W, Ayoub S, Liao J, Sacks MS. A meso‐scale layer‐specific structural constitutive model of the mitral heart valve leaflets. Acta Biomater, 2015.
 300.Zhao B, Etter L, Hinton RB, Jr., Benson DW. BMP and FGF regulatory pathways in semilunar valve precursor cells. Dev Dyn 236: 971‐980, 2007.
 301.Zhou B, von Gise A, Ma Q, Hu YW, Pu WT. Genetic fate mapping demonstrates contribution of epicardium‐derived cells to the annulus fibrosis of the mammalian heart. Dev Biol 338: 251‐261, 2010.

Contact Editor

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

* Required Field

How to Cite

Salma Ayoub, Giovanni Ferrari, Robert C. Gorman, Joseph H. Gorman, Frederick J. Schoen, Michael S. Sacks. Heart Valve Biomechanics and Underlying Mechanobiology. Compr Physiol 2016, 6: 1743-1780. doi: 10.1002/cphy.c150048