1
|
Kampmann C, Lampe C, Wiethoff CM, Arash‐Kaps L, Mengel E, Reinke J, Beck M, Hennermann JB, Abu‐Tair T. Natural history of valve disease in patients with mucopolysaccharidosis II and the impact of enzyme replacement therapy. J Inherit Metab Dis 2025; 48:e12808. [PMID: 39440439 PMCID: PMC11670151 DOI: 10.1002/jimd.12808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Mucopolysaccharidosis II (MPS II, Hunter syndrome) is a rare, X-linked lysosomal storage disease caused by reduced activity of iduronate-2-sulfatase (I2S), with subsequent cellular accumulation of the glycosaminoglycans (GAGs), heparan sulfate, and dermatan sulfate (DS). DS is a major component of the extracellular matrix of heart valves, which can be affected in MPS II. We investigated the natural history of valve disease in MPS II and the impact of long-term intravenous enzyme replacement therapy (ERT) with recombinant I2S (idursulfase). In total, 604 cardiac examinations were assessed from serial follow-up of 80 male patients (49 neuronopathic). Valve disease was classified according to standard practice from hemodynamic features evident from echocardiography. The natural history group comprised 48 patients (up to 14.8 years of follow-up; median, 2.6 years; 24 patients started ERT during the study); 56 patients were treated (up to 14.2 years of follow-up; median, 6.2 years). Lifetime GAG burden (calculated from urinary GAG measurements) correlated significantly with the degree of valve disease. Onset of moderate-to-severe valve disease was significantly delayed in treated (median age at onset, 29.1 ± 2 [95% CI: 25.2-32.9] years; Kaplan-Meier estimation) versus untreated patients (17.6 ± 1 [95% Cl: 15.8-19.4] years; p < 0.0001). Cox regression modeling found that long-term ERT reduced the probability of developing severe valve disease (χ2, 32.736; significant after 5 years of ERT). Overall, this study found that valve disease severity in MPS II correlates with GAG burden and that progression is delayed by long-term ERT.
Collapse
Affiliation(s)
- Christoph Kampmann
- Department of Pediatric Cardiology and Structural Heart Diseases, Center for Diseases in Childhood and AdolescenceUniversity Medicine MainzMainzGermany
| | - Christina Lampe
- Department of Child Neurology, Epileptology and Social MedicineUniversity Hospital GiessenGiessenGermany
| | - Christiane M. Wiethoff
- Department of Pediatric Cardiology and Structural Heart Diseases, Center for Diseases in Childhood and AdolescenceUniversity Medicine MainzMainzGermany
| | | | - Eugen Mengel
- SphinCS, Institute of Clinical Science in LSDHochheimGermany
| | - Joerg Reinke
- Medical center for adults with disabilitiesKreuznacher DiakonieBad KreuznachGermany
| | - Michael Beck
- SphinCS, Institute of Clinical Science in LSDHochheimGermany
| | - Julia B. Hennermann
- Department of Metabolic Diseases, Villa Metabolica, Center for Diseases in Childhood and AdolescenceUniversity Medicine MainzMainzGermany
| | - Tariq Abu‐Tair
- Department of Pediatric Cardiology and Structural Heart Diseases, Center for Diseases in Childhood and AdolescenceUniversity Medicine MainzMainzGermany
| |
Collapse
|
2
|
Parvin Nejad S, Mirani B, Mirzaei Z, Simmons CA. Characterization of pediatric porcine pulmonary valves as a model for tissue engineered heart valves. Acta Biomater 2024; 188:242-252. [PMID: 39233254 DOI: 10.1016/j.actbio.2024.08.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Heart valve tissue engineering holds the potential to transform the surgical management of congenital heart defects affecting the pediatric pulmonary valve (PV) by offering a viable valve replacement. While aiming to recapitulate the native valve, the minimum requirement for tissue engineered heart valves (TEHVs) has historically been adequate mechanical function at implantation. However, long-term in situ functionality of TEHVs remains elusive, suggesting that a closer approximation of the native valve is required. The realization of biomimetic engineered pediatric PV is impeded by insufficient characterization of healthy pediatric tissue. In this study, we comprehensively characterized the planar biaxial tensile behaviour, extracellular matrix (ECM) composition and organization, and valvular interstitial cell (VIC) phenotypes of PVs from piglets to provide benchmarks for TEHVs. The piglet PV possessed an anisotropic and non-linear tension-strain profile from which material constants for a predictive constitutive model were derived. The ECM of the piglet PV possessed a trilayer organization populated by collagen, glycosaminoglycans, and elastin. Biochemical quantification of ECM content normalized to wet weight and DNA content of PV tissue revealed homogeneous distribution across sampled regions of the leaflet. Finally, VICs in the piglet PV were primarily quiescent vimentin-expressing fibroblasts, with a small proportion of activated α-smooth muscle actin-expressing myofibroblasts. Overall, piglet PV properties were consistent with those reported anecdotally for pediatric human PVs and distinct from those of adult porcine and human PVs, supporting the utility of the properties determined here to inform the design of tissue engineered pediatric PVs. STATEMENT OF SIGNIFICANCE: Heart valve tissue engineering has the potential to transform treatment for children born with defective pulmonary valves by providing living replacement tissue that can grow with the child. The design of tissue engineered heart valves is best informed by native valve properties, but native pediatric pulmonary valves have not been fully described to date. Here, we provide comprehensive characterization of the planar biaxial tensile behaviour, extracellular matrix composition and organization, and valvular interstitial cell phenotypes of pulmonary valves from piglets as a model for the native human pediatric valve. Together, these findings provide standards that inform engineered heart valve design towards generation of biomimetic pediatric pulmonary valves.
Collapse
Affiliation(s)
- Shouka Parvin Nejad
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Canada.
| | - Bahram Mirani
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
| | - Zahra Mirzaei
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
| | - Craig A Simmons
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada.
| |
Collapse
|
3
|
Petrovic M, Kahle ER, Han L, Marcolongo MS. Biomimetic proteoglycans as a tool to engineer the structure and mechanics of porcine bioprosthetic heart valves. J Biomed Mater Res B Appl Biomater 2024; 112:e35336. [PMID: 37818847 PMCID: PMC11055403 DOI: 10.1002/jbm.b.35336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/21/2023] [Accepted: 09/18/2023] [Indexed: 10/13/2023]
Abstract
The utility of bioprosthetic heart valves (BHVs) is limited to certain patient populations because of their poor durability compared to mechanical prosthetic valves. Histological analysis of failed porcine BHVs suggests that degeneration of the tissue extracellular matrix (ECM), specifically the loss of proteoglycans and their glycosaminoglycans (GAGs), may lead to impaired mechanical performance, resulting in nucleation and propagation of tears and ultimately failure of the prosthetic. Several strategies have been proposed to address this deterioration, including novel chemical fixatives to stabilize ECM constituents and incorporation of small molecule inhibitors of catabolic enzymes implicated in the degeneration of the BHV ECM. Here, biomimetic proteoglycans (BPGs) were introduced into porcine aortic valves ex vivo and were shown to distribute throughout the valve leaflets. Incorporation of BPGs into the heart valve leaflet increased tissue overall GAG content. The presence of BPGs also significantly increased the micromodulus of the spongiosa layer within the BHV without compromising the chemical fixation process used to sterilize and strengthen the tissue prior to implantation. These findings suggest that a targeted approach for molecularly engineering valve leaflet ECM through the use of BPGs may be a viable way to improve the mechanical behavior and potential durability of BHVs.
Collapse
Affiliation(s)
- Mark Petrovic
- Department of Materials Science and Engineering, Drexel University, Philadelphia, Pennsylvania, USA
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Elizabeth R. Kahle
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Michele S. Marcolongo
- Department of Materials Science and Engineering, Drexel University, Philadelphia, Pennsylvania, USA
- Department of Mechanical Engineering, Villanova University, Villanova, Pennsylvania, USA
| |
Collapse
|
4
|
Thatcher K, Mattern CR, Chaparro D, Goveas V, McDermott MR, Fulton J, Hutcheson JD, Hoffmann BR, Lincoln J. Temporal Progression of Aortic Valve Pathogenesis in a Mouse Model of Osteogenesis Imperfecta. J Cardiovasc Dev Dis 2023; 10:355. [PMID: 37623368 PMCID: PMC10455328 DOI: 10.3390/jcdd10080355] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Organization of extracellular matrix (ECM) components, including collagens, proteoglycans, and elastin, is essential for maintaining the structure and function of heart valves throughout life. Mutations in ECM genes cause connective tissue disorders, including Osteogenesis Imperfecta (OI), and progressive debilitating heart valve dysfunction is common in these patients. Despite this, effective treatment options are limited to end-stage interventions. Mice with a homozygous frameshift mutation in col1a2 serve as a murine model of OI (oim/oim), and therefore, they were used in this study to examine the pathobiology of aortic valve (AoV) disease in this patient population at structural, functional, and molecular levels. Temporal echocardiography of oim/oim mice revealed AoV dysfunction by the late stages of disease in 12-month-old mice. However, structural and proteomic changes were apparent much earlier, at 3 months of age, and were associated with disturbances in ECM homeostasis primarily related to collagen and proteoglycan abnormalities and disorganization. Together, findings from this study provide insights into the underpinnings of late onset AoV dysfunction in connective tissue disease patients that can be used for the development of mechanistic-based therapies administered early to halt progression, thereby avoiding late-stage surgical intervention.
Collapse
Affiliation(s)
- Kaitlyn Thatcher
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (K.T.); (C.R.M.); (V.G.)
- Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - Carol R. Mattern
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (K.T.); (C.R.M.); (V.G.)
- Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - Daniel Chaparro
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (D.C.); (J.D.H.)
| | - Veronica Goveas
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (K.T.); (C.R.M.); (V.G.)
- Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - Michael R. McDermott
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA; (M.R.M.); (J.F.)
| | - Jessica Fulton
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA; (M.R.M.); (J.F.)
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA; (D.C.); (J.D.H.)
| | - Brian R. Hoffmann
- Mass Spectrometry and Protein Chemistry, Protein Sciences, The Jackson Laboratory, Bar Harbor, ME 04609, USA;
| | - Joy Lincoln
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (K.T.); (C.R.M.); (V.G.)
- Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
5
|
Liu Y, Chen C, Lu T, Liu S, Wu Z, Tang Z. Free-aldehyde neutralized and oligohyaluronan loaded bovine pericardium with improved anti-calcification and endothelialization for bioprosthetic heart valves. Front Bioeng Biotechnol 2023; 11:1138972. [PMID: 37077226 PMCID: PMC10106738 DOI: 10.3389/fbioe.2023.1138972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
The number of patients with valvular heart disease is increasing yearly, and valve replacement is the most effective treatment, during which bioprosthetic heart valves (BHVs) are the most widely used. Commercial BHVs are mainly prepared with glutaraldehyde (Glut) cross-linked bovine pericardial or porcine aortic valves, but the residual free aldehyde groups in these tissues can cause calcification and cytotoxicity. Moreover, insufficient glycosaminoglycans (GAGs) in tissues can further reduce biocompatibility and durability. However, the anti-calcification performance and biocompatibility might be improved by blocking the free aldehyde groups and increasing the GAGs content in Glut-crosslinked tissues. In our study, adipic dihydrazide (ADH) was used to neutralize the residual free aldehyde groups in tissues and provide sites to blind with oligohyaluronan (OHA) to increase the content of GAGs in tissues. The modified bovine pericardium was evaluated for its content of residual aldehyde groups, the amount of OHA loaded, physical/chemical characteristics, biomechanical properties, biocompatibility, and in vivo anticalcification assay and endothelialization effects in juvenile Sprague-Dawley rats. The results showed that ADH could completely neutralize the free aldehyde groups in the Glut-crosslinked bovine pericardium, the amount of OHA loaded increased and the cytotoxicity was reduced. Moreover, the in vivo results also showed that the level of calcification and inflammatory response in the modified pericardial tissue was significantly reduced in a rat subcutaneous implantation model, and the results from the rat abdominal aorta vascular patch repair model further demonstrated the improved capability of the modified pericardial tissues for endothelialization. Furthermore, more α-SMA+ smooth muscle cells and fewer CD68+ macrophages infiltrated in the neointima of the modified pericardial patch. In summary, blocking free-aldehydes and loading OHA improved the anti-calcification, anti-inflammation and endothelialization properties of Glut-crosslinked BHVs and in particularly, this modified strategy may be a promising candidate for the next-generation of BHVs.
Collapse
Affiliation(s)
- Yuhong Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering Laboratory of Human Province for Cardiovascular Biomaterials, Changsha, Hunan, China
| | - Chunyang Chen
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering Laboratory of Human Province for Cardiovascular Biomaterials, Changsha, Hunan, China
| | - Ting Lu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering Laboratory of Human Province for Cardiovascular Biomaterials, Changsha, Hunan, China
| | - Sixi Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering Laboratory of Human Province for Cardiovascular Biomaterials, Changsha, Hunan, China
| | - Zhongshi Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering Laboratory of Human Province for Cardiovascular Biomaterials, Changsha, Hunan, China
- *Correspondence: Zhongshi Wu, ; Zhenjie Tang,
| | - Zhenjie Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering Laboratory of Human Province for Cardiovascular Biomaterials, Changsha, Hunan, China
- *Correspondence: Zhongshi Wu, ; Zhenjie Tang,
| |
Collapse
|
6
|
Wu L, Huang K, Li Q, Wu H, Gao Y, Xu X, Liu X, Han L. Crosstalk between myofibroblasts and macrophages: A regulative factor of valvular fibrosis in calcific aortic valve disease. Cell Biol Int 2023; 47:754-767. [PMID: 36542640 DOI: 10.1002/cbin.11980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Inflammation and fibrosis are highly correlated with the progression of calcific aortic valve disease (CAVD). As one of the differentiated forms of valvular interstitial cells, myofibroblasts play a critical role in CAVD's development as do macrophages. Although numerous studies have been conducted on them separately, their communication and interaction remain unclear. We used porcine aortic valves to isolate valve interstitial cells (VICs). VICs were induced to differentiate into myofibroblasts by transforming growth factor-β1 (TGF-β1). After successful activation was determined, the myofibroblast-conditioned medium (CM) was collected and used to act on RAW264.7, a macrophage cell line. A migration and adhesion assay estimated the recruitment capability of myofibroblasts on macrophages. We used flow cytometry, quantitative polymerase chain reaction (qPCR), and Western blot analysis to investigate myofibroblasts' polarity promotion function in macrophages. Finally, we used macrophage-CM on VICs to explore the differentiation induction function of polarized macrophages. Myofibroblast marker alpha-smooth muscle actin and M2 macrophage marker CD163 were detected as upregulated in CAVD patients, and their expression has a certain correlation. The Smad3/HA/CD44 axis activated the differentiation of myofibroblasts by Western blot. The myofibroblast-CM can promote chemotaxis and adhesion of macrophages through protein kinase B/chemokine (C-C motif) ligand5 and Smad3/HA/CD44, respectively. Hyaluronic acid (HA) inside the myofibroblast-CM stimulates macrophages to polarize into M2 macrophages. In turn, M2 macrophage-CM has the promotive ability to activate myofibroblasts but fails to induce the osteoblast differentiation of VICs directly. The crosstalk between myofibroblasts and macrophages causes the excessive activation of myofibroblasts. This positive feedback loop may play a vital role in CAVD progression.
Collapse
Affiliation(s)
- Lujia Wu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Kai Huang
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qin Li
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hao Wu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuan Gao
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiangyang Xu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaohong Liu
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lin Han
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
7
|
Ingeniería de tejidos en población pediátrica: una esperanza para el tratamiento de enfermedades valvulares mitrales congénitas. CIRUGIA CARDIOVASCULAR 2023. [DOI: 10.1016/j.circv.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
8
|
Bouten CVC, Cheng C, Vermue IM, Gawlitta D, Passier R. Cardiovascular tissue engineering and regeneration: A plead for further knowledge convergence. Tissue Eng Part A 2022; 28:525-541. [PMID: 35382591 DOI: 10.1089/ten.tea.2021.0231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular tissue engineering and regeneration strive to provide long-term, effective solutions for a growing group of patients in need of myocardial repair, vascular (access) grafts, heart valves, and regeneration of organ microcirculation. In the past two decades, ongoing convergence of disciplines and multidisciplinary collaborations between cardiothoracic surgeons, cardiologists, bioengineers, material scientists, and cell biologists have resulted in better understanding of the problems at hand and novel regenerative approaches. As a side effect, however, the field has become strongly organized and differentiated around topical areas at risk of reinvention of technologies and repetition of approaches and across the areas. A better integration of knowledge and technologies from the individual topical areas and regenerative approaches and technologies may pave the way towards faster and more effective treatments to cure the cardiovascular system. This review summarizes the evolution of research and regenerative approaches in the areas of myocardial regeneration, heart valve and vascular tissue engineering, and regeneration of microcirculations and discusses previous and potential future integration of these individual areas and developed technologies for improved clinical impact. Finally, it provides a perspective on the further integration of research organization, knowledge implementation, and valorization as a contributor to advancing cardiovascular tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Carlijn V C Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Caroline Cheng
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Experimental Cardiology, Department of Cardiology, Thoraxcenter Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ijsbrand M Vermue
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery, Prosthodontics and Special Dental Care, University Medical Center, Utrecht, The Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
9
|
Passos LSA, Becker-Greene D, Braulio R, Le TD, Gelape CL, de Almeida LFR, Rocha DPA, Gomes CAP, Esteves WAM, Passaglia LG, Dal-Bianco JP, Levine RA, Aikawa M, Hung J, Dutra WO, Nunes MCP, Aikawa E. Proinflammatory Matrix Metalloproteinase-1 Associates With Mitral Valve Leaflet Disruption Following Percutaneous Mitral Valvuloplasty. Front Cardiovasc Med 2022; 8:804111. [PMID: 35127864 PMCID: PMC8811173 DOI: 10.3389/fcvm.2021.804111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022] Open
Abstract
Mitral regurgitation (MR) is a major complication of the percutaneous mitral valvuloplasty (PMV). Despite high technical expertise and cumulative experience with the procedure, the incidence rate of severe MR has not decreased. Although some of MR can be anticipated by echocardiographic analysis; leaflet tearing, which leads to the most dreaded type of MR, remains unpredictable. Irregular valvular collagen remodeling is likely to compromise tissue architecture and increase the tearing risk during PMV balloon inflation. In this study, we evaluated histological and molecular characteristics of excised mitral valves from patients with rheumatic mitral stenosis (MS) who underwent emergency surgery after PMV due to severe MR caused by leaflet tear. Those findings were compared with patients who underwent elective mitral valve replacement surgery owing to severe MS, in whom PMV was not indicated. In vitro assay using peripheral blood mononuclear cells was performed to better understand the impact of the cellular and molecular alterations identified in leaflet tear mitral valve specimens. Our analysis showed that focal infiltration of inflammatory cells contributes to accumulation of MMP-1 and IFN-γ in valve leaflets. Moreover, we showed that IFN-γ increase the expression of MMP-1 in CD14+ cells (monocytes) in vitro. Thus, inflammatory cells contribute to unevenly remodel collagen resulting in variable thickening causing abnormalities in leaflet architecture making them more susceptible to laceration.
Collapse
Affiliation(s)
- Livia S. A. Passos
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Dakota Becker-Greene
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Renato Braulio
- School of Medicine, Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Thanh-Dat Le
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Cláudio L. Gelape
- School of Medicine, Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luís Felipe R. de Almeida
- School of Medicine, Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Divino Pedro A. Rocha
- School of Medicine, Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carlos Augusto P. Gomes
- School of Medicine, Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - William A. M. Esteves
- School of Medicine, Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luiz G. Passaglia
- School of Medicine, Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jacob P. Dal-Bianco
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Robert A. Levine
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Judy Hung
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Walderez O. Dutra
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Belo Horizonte, Brazil
| | - Maria Carmo P. Nunes
- School of Medicine, Hospital das Clínicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
- *Correspondence: Elena Aikawa
| |
Collapse
|
10
|
Engineering the aortic valve extracellular matrix through stages of development, aging, and disease. J Mol Cell Cardiol 2021; 161:1-8. [PMID: 34339757 DOI: 10.1016/j.yjmcc.2021.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 02/01/2023]
Abstract
For such a thin tissue, the aortic valve possesses an exquisitely complex, multi-layered extracellular matrix (ECM), and disruptions to this structure constitute one of the earliest hallmarks of fibrocalcific aortic valve disease (CAVD). The native valve structure provides a challenging target for engineers to mimic, but the development of advanced, ECM-based scaffolds may enable mechanistic and therapeutic discoveries that are not feasible in other culture or in vivo platforms. This review first discusses the ECM changes that occur during heart valve development, normal aging, onset of early-stage disease, and progression to late-stage disease. We then provide an overview of the bottom-up tissue engineering strategies that have been used to mimic the valvular ECM, and opportunities for advancement in these areas.
Collapse
|
11
|
Shao Z, Tao T, Xu H, Chen C, Lee I, Chung S, Dong Z, Li W, Ma L, Bai H, Chen Q. Recent progress in biomaterials for heart valve replacement: Structure, function, and biomimetic design. VIEW 2021. [DOI: 10.1002/viw.20200142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Ziyu Shao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine & Clinical Research Center for Oral Diseases of Zhejiang Province Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University Hangzhou 310006 China
- State Key Laboratory of Chemical Engineering College of Chemical and Biological Engineering Zhejiang University Hangzhou China
| | - Tingting Tao
- Department of Cardiovascular Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang Province China
| | - Hongfei Xu
- Department of Cardiovascular Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang Province China
| | - Cen Chen
- College of Life Sciences and Medicine Zhejiang Sci‐Tech University Hangzhou China
| | - In‐Seop Lee
- College of Life Sciences and Medicine Zhejiang Sci‐Tech University Hangzhou China
- Institute of Natural Sciences Yonsei University Seoul Republic of Korea
| | - Sungmin Chung
- Biomaterials R&D Center GENOSS Co., Ltd. Suwon‐si Republic of Korea
| | - Zhihui Dong
- State Key Laboratory of Chemical Engineering College of Chemical and Biological Engineering Zhejiang University Hangzhou China
| | - Weidong Li
- Department of Cardiovascular Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang Province China
| | - Liang Ma
- Department of Cardiovascular Surgery The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang Province China
| | - Hao Bai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine & Clinical Research Center for Oral Diseases of Zhejiang Province Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University Hangzhou 310006 China
- State Key Laboratory of Chemical Engineering College of Chemical and Biological Engineering Zhejiang University Hangzhou China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine & Clinical Research Center for Oral Diseases of Zhejiang Province Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University Hangzhou 310006 China
| |
Collapse
|
12
|
Spatial N-glycomics of the human aortic valve in development and pediatric endstage congenital aortic valve stenosis. J Mol Cell Cardiol 2021; 154:6-20. [PMID: 33516683 DOI: 10.1016/j.yjmcc.2021.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022]
Abstract
Congenital aortic valve stenosis (AS) progresses as an obstructive narrowing of the aortic orifice due to deregulated extracellular matrix (ECM) production by aortic valve (AV) leaflets and leads to heart failure with no effective therapies. Changes in glycoprotein and proteoglycan distribution are a hallmark of AS, yet valvular carbohydrate content remains virtually uncharacterized at the molecular level. While almost all glycoproteins clinically linked to stenotic valvular modeling contain multiple sites for N-glycosylation, there are very few reports aimed at understanding how N-glycosylation contributes to the valve structure in disease. Here, we tested for spatial localization of N-glycan structures within pediatric congenital aortic valve stenosis. The study was done on valvular tissues 0-17 years of age with de-identified clinical data reporting pre-operative valve function spanning normal development, aortic valve insufficiency (AVI), and pediatric endstage AS. High mass accuracy imaging mass spectrometry (IMS) was used to localize N-glycan profiles in the AV structure. RNA-Seq was used to identify regulation of N-glycan related enzymes. The N-glycome was found to be spatially localized in the normal aortic valve, aligning with fibrosa, spongiosa or ventricularis. In AVI diagnosed tissue, N-glycans localized to hypertrophic commissures with increases in pauci-mannose structures. In all valve types, sialic acid (N-acetylneuraminic acid) N-glycans were the most abundant N-glycan group. Three sialylated N-glycans showed common elevation in AS independent of age. On-tissue chemical methods optimized for valvular tissue determined that aortic valve tissue sialylation shows both α2,6 and α2,3 linkages. Specialized enzymatic strategies demonstrated that core fucosylation is the primary fucose configuration and localizes to the normal fibrosa with disparate patterning in AS. This study identifies that the human aortic valve structure is spatially defined by N-glycomic signaling and may generate new research directions for the treatment of human aortic valve disease.
Collapse
|
13
|
Extracellular Matrix in Calcific Aortic Valve Disease: Architecture, Dynamic and Perspectives. Int J Mol Sci 2021; 22:ijms22020913. [PMID: 33477599 PMCID: PMC7831300 DOI: 10.3390/ijms22020913] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/18/2022] Open
Abstract
Calcific Aortic Valve Disease (CAVD) is the most common valvular heart disease in developed countries and in the ageing population. It is strongly correlated to median age, affecting up to 13% of the population over the age of 65. Pathophysiological analysis indicates CAVD as a result of an active and degenerative disease, starting with sclerosis and chronic inflammation and then leaflet calcification, which ultimately can account for aortic stenosis. Although CAVD has been firstly recognized as a passive event mostly resulting from a degenerative aging process, much evidences suggests that calcification arises from different active processes, involving both aortic valve-resident cells (valve endothelial cells, valve interstitial cells, mesenchymal stem cells, innate immunity cells) and circulating cells (circulating mesenchymal cells, immunity cells). Moreover, a role for the cell-derived "matrix vesicles" and extracellular matrix (ECM) components has also been recognized. The aim of this work is to review the cellular and molecular alterations occurring in aortic valve during CAVD pathogenesis, focusing on the role of ECM in the natural course of the disease.
Collapse
|
14
|
Biology and Biomechanics of the Heart Valve Extracellular Matrix. J Cardiovasc Dev Dis 2020; 7:jcdd7040057. [PMID: 33339213 PMCID: PMC7765611 DOI: 10.3390/jcdd7040057] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/02/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023] Open
Abstract
Heart valves are dynamic structures that, in the average human, open and close over 100,000 times per day, and 3 × 109 times per lifetime to maintain unidirectional blood flow. Efficient, coordinated movement of the valve structures during the cardiac cycle is mediated by the intricate and sophisticated network of extracellular matrix (ECM) components that provide the necessary biomechanical properties to meet these mechanical demands. Organized in layers that accommodate passive functional movements of the valve leaflets, heart valve ECM is synthesized during embryonic development, and remodeled and maintained by resident cells throughout life. The failure of ECM organization compromises biomechanical function, and may lead to obstruction or leaking, which if left untreated can lead to heart failure. At present, effective treatment for heart valve dysfunction is limited and frequently ends with surgical repair or replacement, which comes with insuperable complications for many high-risk patients including aged and pediatric populations. Therefore, there is a critical need to fully appreciate the pathobiology of biomechanical valve failure in order to develop better, alternative therapies. To date, the majority of studies have focused on delineating valve disease mechanisms at the cellular level, namely the interstitial and endothelial lineages. However, less focus has been on the ECM, shown previously in other systems, to be a promising mechanism-inspired therapeutic target. Here, we highlight and review the biology and biomechanical contributions of key components of the heart valve ECM. Furthermore, we discuss how human diseases, including connective tissue disorders lead to aberrations in the abundance, organization and quality of these matrix proteins, resulting in instability of the valve infrastructure and gross functional impairment.
Collapse
|
15
|
Wu S, Kumar V, Xiao P, Kuss M, Lim JY, Guda C, Butcher J, Duan B. Age related extracellular matrix and interstitial cell phenotype in pulmonary valves. Sci Rep 2020; 10:21338. [PMID: 33288823 PMCID: PMC7721746 DOI: 10.1038/s41598-020-78507-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/24/2020] [Indexed: 12/21/2022] Open
Abstract
Heart valve disease is a common manifestation of cardiovascular disease and is a significant cause of cardiovascular morbidity and mortality worldwide. The pulmonary valve (PV) is of primary concern because of its involvement in common congenital heart defects, and the PV is usually the site for prosthetic replacement following a Ross operation. Although effects of age on valve matrix components and mechanical properties for aortic and mitral valves have been studied, very little is known about the age-related alterations that occur in the PV. In this study, we isolated PV leaflets from porcine hearts in different age groups (~ 4-6 months, denoted as young versus ~ 2 years, denoted as adult) and studied the effects of age on PV leaflet thickness, extracellular matrix components, and mechanical properties. We also conducted proteomics and RNA sequencing to investigate the global changes of PV leaflets and passage zero PV interstitial cells in their protein and gene levels. We found that the size, thickness, elastic modulus, and ultimate stress in both the radial and circumferential directions and the collagen of PV leaflets increased from young to adult age, while the ultimate strain and amount of glycosaminoglycans decreased when age increased. Young and adult PV had both similar and distinct protein and gene expression patterns that are related to their inherent physiological properties. These findings are important for us to better understand the physiological microenvironments of PV leaflet and valve cells for correctively engineering age-specific heart valve tissues.
Collapse
Affiliation(s)
- Shaohua Wu
- College of Textiles & Clothing, Qingdao University, Qingdao, People's Republic of China
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vikas Kumar
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peng Xiao
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jonathan Butcher
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA.
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
16
|
Mechanosensing dysregulation in the fibroblast: A hallmark of the aging heart. Ageing Res Rev 2020; 63:101150. [PMID: 32846223 DOI: 10.1016/j.arr.2020.101150] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
The myofibroblast is a specialized fibroblast that expresses α-smooth muscle actin (α-SMA) and participates in wound contraction and fibrosis. The fibroblast to myofibroblast transition depends on chemical and mechanical signals. A fibroblast senses the changes in the environment (extracellular matrix (ECM)) and transduces these changes to the cytoskeleton and the nucleus, resulting in activation or inhibition of α-SMA transcription in a process called mechanosensing. A stiff matrix greatly facilitates the transition from fibroblast to myofibroblast, and although the aging heart is much stiffer than the young one, the aging fibroblast has difficulties in transitioning into the contractile phenotype. This suggests that the events occurring downstream of the matrix, such as activation or changes in expression levels of various proteins participating in mechanotransduction can negatively alter the ability of the aging fibroblast to become a myofibroblast. In this review, we will discuss in detail the changes in ECM, receptors (integrin or non-integrin), focal adhesions, cytoskeleton, and transcription factors involved in mechanosensing that occur with aging.
Collapse
|
17
|
Madhurapantula RS, Krell G, Morfin B, Roy R, Lister K, Orgel JP. Advanced Methodology and Preliminary Measurements of Molecular and Mechanical Properties of Heart Valves under Dynamic Strain. Int J Mol Sci 2020; 21:E763. [PMID: 31991583 PMCID: PMC7037596 DOI: 10.3390/ijms21030763] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 11/16/2022] Open
Abstract
Mammalian heart valves are soft tissue assemblies with multi-scale material properties. This is because they are constructs comprising both muscle and non-contractile extracellular matrix proteins (such as collagens and proteoglycans) and transition regions where one form of tissue structure becomes another, significantly different form. The leaflets of the mitral and tricuspid valves are connected to chordae tendinae which, in turn, bind through papillary muscles to the cardiac wall of the ventricle. The transition regions between these tissue subsets are complex and diffuse. Their material composition and mechanical properties have not been previously described with both micro and nanoscopic data recorded simultaneously, as reported here. Annotating the mechanical characteristics of these tissue transitions will be of great value in developing novel implants, improving the state of the surgical simulators and advancing robot-assisted surgery. We present here developments in multi-scale methodology that produce data that can relate mechanical properties to molecular structure using scanning X-ray diffraction. We correlate these data to corresponding tissue level (macro and microscopic) stress and strain, with particular emphasis on the transition regions and present analyses to indicate points of possible failure in these tissues.
Collapse
Affiliation(s)
- Rama S. Madhurapantula
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA;
- Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA;
| | - Gabriel Krell
- Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA;
| | - Berenice Morfin
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA;
| | - Rajarshi Roy
- Corvid Technologies, Mooresville, NC 28117, USA; (R.R.); (K.L.)
| | - Kevin Lister
- Corvid Technologies, Mooresville, NC 28117, USA; (R.R.); (K.L.)
| | - Joseph P.R.O. Orgel
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA;
- Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
18
|
Oyama MA, Elliott C, Loughran KA, Kossar AP, Castillero E, Levy RJ, Ferrari G. Comparative pathology of human and canine myxomatous mitral valve degeneration: 5HT and TGF-β mechanisms. Cardiovasc Pathol 2020; 46:107196. [PMID: 32006823 DOI: 10.1016/j.carpath.2019.107196] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/16/2019] [Accepted: 12/28/2019] [Indexed: 12/25/2022] Open
Abstract
Myxomatous mitral valve degeneration (MMVD) is a leading cause of valve repair or replacement secondary to the production of mitral regurgitation, cardiac enlargement, systolic dysfunction, and heart failure. The pathophysiology of myxomatous mitral valve degeneration is complex and incompletely understood, but key features include activation and transformation of mitral valve (MV) valvular interstitial cells (VICs) into an active phenotype leading to remodeling of the extracellular matrix and compromise of the structural components of the mitral valve leaflets. Uncovering the mechanisms behind these events offers the potential for therapies to prevent, delay, or reverse myxomatous mitral valve degeneration. One such mechanism involves the neurotransmitter serotonin (5HT), which has been linked to development of valvulopathy in a variety of settings, including valvulopathy induced by serotonergic drugs, Serotonin-producing carcinoid tumors, and development of valvulopathy in laboratory animals exposed to high levels of serotonin. Similar to humans, the domestic dog also experiences naturally occurring myxomatous mitral valve degeneration, and in some breeds of dogs, the lifetime prevalence of myxomatous mitral valve degeneration reaches 100%. In dogs, myxomatous mitral valve degeneration has been associated with high serum serotonin, increased expression of serotonin-receptors, autocrine production of serotonin within the mitral valve leaflets, and downregulation of serotonin clearance mechanisms. One pathway closely associated with serotonin involves transforming growth factor beta (TGF-β) and the two pathways share a common ability to activate mitral valve valvular interstitial cells in both humans and dogs. Understanding the role of serotonin and transforming growth factor beta in myxomatous mitral valve degeneration gives rise to potential therapies, such as 5HT receptor (5HT-R) antagonists. The main purposes of this review are to highlight the commonalities between myxomatous mitral valve degeneration in humans and dogs, with specific regards to serotonin and transforming growth factor beta, and to champion the dog as a relevant and particularly valuable model of human disease that can accelerate development of novel therapies.
Collapse
Affiliation(s)
- Mark A Oyama
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chad Elliott
- Department of Surgery, Columbia Cardiovascular Institute and College of Physicians and Surgeons at Columbia University, New York, NY, USA
| | - Kerry A Loughran
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander P Kossar
- Department of Surgery, Columbia Cardiovascular Institute and College of Physicians and Surgeons at Columbia University, New York, NY, USA
| | - Estibaliz Castillero
- Department of Surgery, Columbia Cardiovascular Institute and College of Physicians and Surgeons at Columbia University, New York, NY, USA
| | - Robert J Levy
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Giovanni Ferrari
- Department of Surgery, Columbia Cardiovascular Institute and College of Physicians and Surgeons at Columbia University, New York, NY, USA.
| |
Collapse
|
19
|
Kristen M, Ainsworth MJ. Fiber Scaffold Patterning for Mending Hearts: 3D Organization Bringing the Next Step. Adv Healthc Mater 2020; 9:e1900775. [PMID: 31603288 PMCID: PMC7116178 DOI: 10.1002/adhm.201900775] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/14/2019] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) is a leading cause of death worldwide. The most common conditions that lead to HF are coronary artery disease, myocardial infarction, valve disorders, high blood pressure, and cardiomyopathy. Due to the limited regenerative capacity of the heart, the only curative therapy currently available is heart transplantation. Therefore, there is a great need for the development of novel regenerative strategies to repair the injured myocardium, replace damaged valves, and treat occluded coronary arteries. Recent advances in manufacturing technologies have resulted in the precise fabrication of 3D fiber scaffolds with high architectural control that can support and guide new tissue growth, opening exciting new avenues for repair of the human heart. This review discusses the recent advancements in the novel research field of fiber patterning manufacturing technologies for cardiac tissue engineering (cTE) and to what extent these technologies could meet the requirements of the highly organized and structured cardiac tissues. Additionally, future directions of these novel fiber patterning technologies, designs, and applicability to advance cTE are presented.
Collapse
Affiliation(s)
- Marleen Kristen
- Regenerative Medicine Center, University Medical Center Utrecht,
Utrecht 3584 CT, The Netherlands; Department of Orthopedics, University Medical
Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Madison J. Ainsworth
- Regenerative Medicine Center, University Medical Center Utrecht,
Utrecht 3584 CT, The Netherlands; Department of Orthopedics, University Medical
Center Utrecht, Utrecht 3584 CX, The Netherlands
| |
Collapse
|
20
|
Contrast enhanced computed tomography for real-time quantification of glycosaminoglycans in cartilage tissue engineered constructs. Acta Biomater 2019; 100:202-212. [PMID: 31580960 DOI: 10.1016/j.actbio.2019.09.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022]
Abstract
Tissue engineering and regenerative medicine are two therapeutic strategies to treat, and to potentially cure, diseases affecting cartilaginous tissues, such as osteoarthritis and cartilage defects. Insights into the processes occurring during regeneration are essential to steer and inform development of the envisaged regenerative strategy, however tools are needed for longitudinal and quantitative monitoring of cartilage matrix components. In this study, we introduce a contrast-enhanced computed tomography (CECT)-based method using a cationic iodinated contrast agent (CA4+) for longitudinal quantification of glycosaminoglycans (GAG) in cartilage-engineered constructs. CA4+ concentration and scanning protocols were first optimized to ensure no cytotoxicity and a facile procedure with minimal radiation dose. Chondrocyte and mesenchymal stem cell pellets, containing different GAG content were generated and exposed to CA4+. The CA4+ content in the pellets, as determined by micro computed tomography, was plotted against GAG content, as measured by 1,9-dimethylmethylene blue analysis, and showed a high linear correlation. The established equation was used for longitudinal measurements of GAG content over 28 days of pellet culture. Importantly, this method did not adversely affect cell viability or chondrogenesis. Additionally, the CA4+ distribution accurately matched safranin-O staining on histological sections. Hence, we show proof-of-concept for the application of CECT, utilizing a positively charged contrast agent, for longitudinal and quantitative imaging of GAG distribution in cartilage tissue-engineered constructs. STATEMENT OF SIGNIFICANCE: Tissue engineering and regenerative medicine are promising therapeutic strategies for different joint pathologies such as cartilage defects or osteoarthritis. Currently, in vitro assessment on the quality and composition of the engineered cartilage mainly relies on destructive methods. Therefore, there is a need for the development of techniques that allow for longitudinal and quantitative imaging and monitoring of cartilage-engineered constructs. This work harnesses the electrostatic interactions between the negatively-charged glycosaminoglycans (GAGs) and a positively-charged contrast agent for longitudinal and non-destructive quantification of GAGs, providing valuable insight on GAG development and distribution in cartilage engineered constructs. Such technique can advance the development of regenerative strategies, not only by allowing continuous monitoring but also by serving as a pre-implantation screening tool.
Collapse
|
21
|
Vekilov DP, Singh M, Aglyamov SR, Larin KV, Grande-Allen KJ. Mapping the spatial variation of mitral valve elastic properties using air-pulse optical coherence elastography. J Biomech 2019; 93:52-59. [PMID: 31300156 PMCID: PMC10575695 DOI: 10.1016/j.jbiomech.2019.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/18/2019] [Accepted: 06/14/2019] [Indexed: 10/26/2022]
Abstract
The mitral valve is a highly heterogeneous tissue composed of two leaflets, anterior and posterior, whose unique composition and regional differences in material properties are essential to overall valve function. While mitral valve mechanics have been studied for many decades, traditional testing methods limit the spatial resolution of measurements and can be destructive. Optical coherence elastography (OCE) is an emerging method for measuring viscoelastic properties of tissues in a noninvasive, nondestructive manner. In this study, we employed air-pulse OCE to measure the spatial variation in mitral valve elastic properties with micro-scale resolution at 1 mm increments along the radial length of the leaflets. We analyzed differences between the leaflets, as well as between regions of the valve. We found that the anterior leaflet has a higher elastic wave velocity, which is reported as a surrogate for stiffness, than the posterior leaflet, most notably at the annular edge of the sample. In addition, we found a spatial elastic gradient in the anterior leaflet, where the annular edge was found to have a greater elastic wave velocity than the free edge. This gradient was less pronounced in the posterior leaflet. These patterns were confirmed using established uniaxial tensile testing methods. Overall, the anterior leaflet was stiffer and had greater heterogeneity in its mechanical properties than the posterior leaflet. This study measures differences between the two mitral leaflets with greater resolution than previously feasible and demonstrates a method that may be suitable for assessing valve mechanics following repair or during the engineering of synthetic valve replacements.
Collapse
Affiliation(s)
| | - Manmohan Singh
- University of Houston, Department of Biomedical Engineering, Houston, TX, United States
| | - Salavat R Aglyamov
- University of Houston, Department of Mechanical Engineering, Houston, TX, United States; University of Texas at Austin, Department of Biomedical Engineering, Austin, TX, United States
| | - Kirill V Larin
- University of Houston, Department of Biomedical Engineering, Houston, TX, United States
| | | |
Collapse
|
22
|
Baugh L, Watson MC, Kemmerling EC, Hinds PW, Huggins GS, Black LD. Knockdown of CD44 expression decreases valve interstitial cell calcification in vitro. Am J Physiol Heart Circ Physiol 2019; 317:H26-H36. [PMID: 30951363 PMCID: PMC6692733 DOI: 10.1152/ajpheart.00123.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/23/2022]
Abstract
The lack of pharmaceutical targets available to treat patients with calcific aortic valve disease (CAVD) necessitates further research into the specific mechanisms of the disease. The significant changes that occur to the aortic valves extracellular matrix (ECM) during the progression of CAVD suggests that these proteins may play an important role in calcification. Exploring the relationship between valve interstitial cells (VICs) and the ECM may lead to a better understand of CAVD mechanisms and potential pharmaceutical targets. In this study, we look at the effect of two ECM components, collagen and hyaluronic acid (HA), on the mineralization of VICs within the context of a two-dimensional, polyacrylamide (PAAM) model system. Using a novel, nondestructive imaging technique, we were able to track calcific nodule development in culture systems over a 3-wk time frame. We saw a significant increase in the size of the nodules grown on HA PAAM gels as compared with collagen PAAM gels, suggesting that HA has a direct effect on mineralization. Directly looking at the two known receptors of HA, CD44 and receptor for HA-mediated motility (RHAMM), and using siRNA knockdown revealed that a decrease in CD44 expression resulted in a reduction of calcification. A decrease in CD44, through siRNA knockdown, reduces mineralization on HA PAAM gels, suggesting a potential new target for CAVD treatment. NEW & NOTEWORTHY Our in vitro model of calcific aortic valve disease shows an interaction between the hyaluronic acid binding protein CD44 with the osteogenic factor OPN as a potential mechanism of aortic valve calcification. Using siRNA knockdown of CD44, we show an upregulation of OPN expression with a decrease in overall mineralization.
Collapse
Affiliation(s)
- Lauren Baugh
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
| | - Matthew C Watson
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
- Department of Mechanical Engineering, Tufts University , Medford, Massachusetts
| | - Erica C Kemmerling
- Department of Mechanical Engineering, Tufts University , Medford, Massachusetts
| | - Philip W Hinds
- Cellular, Molecular, and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine , Boston, Massachusetts
| | - Gordon S Huggins
- Molecular Cardiology Research Center, Tufts Medical Center and Tufts University Sackler School for Graduate Biomedical Sciences , Boston, Massachusetts
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine , Boston, Massachusetts
| |
Collapse
|
23
|
Barth M, Selig JI, Klose S, Schomakers A, Kiene LS, Raschke S, Boeken U, Akhyari P, Fischer JW, Lichtenberg A. Degenerative aortic valve disease and diabetes: Implications for a link between proteoglycans and diabetic disorders in the aortic valve. Diab Vasc Dis Res 2019; 16:254-269. [PMID: 30563371 DOI: 10.1177/1479164118817922] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Degenerative aortic valve disease in combination with diabetes is an increasing burden worldwide. There is growing evidence that particularly small leucine-rich proteoglycans are involved in the development of degenerative aortic valve disease. Nevertheless, the role of these molecules in this disease in the course of diabetes has not been elucidated in detail and previous studies remain controversial. Therefore, the aim of this study is to broaden the knowledge about small leucine-rich proteoglycans in degenerative aortic valve disease and the influence of diabetes and hyperglycaemia on aortic valves and valvular interstitial cells is examined. Analyses were performed using reverse-transcription polymerase chain reaction, Western blot, enzyme-linked immunosorbent assay, (immuno)histology and colorimetric assays. We could show that biglycan, but not decorin and lumican, is upregulated in degenerated human aortic valve cusps. Subgroup analysis reveals that upregulation of biglycan is stage-dependent. In vivo, loss of biglycan leads to stage-dependent calcification and also to migratory effects on interstitial cells within the extracellular matrix. In late stages of degenerative aortic valve disease, diabetes increases the expression of biglycan in aortic valves. In vitro, the combinations of hyperglycaemic with pro-degenerative conditions lead to an upregulation of biglycan. In conclusion, biglycan represents a potential link between degenerative aortic valve disease and diabetes.
Collapse
Affiliation(s)
- Mareike Barth
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jessica I Selig
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Svenja Klose
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Antje Schomakers
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Lena S Kiene
- 2 Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Silja Raschke
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Udo Boeken
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Payam Akhyari
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jens W Fischer
- 2 Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Artur Lichtenberg
- 1 Department of Cardiovascular Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
24
|
Jana S, Lerman A. Behavior of valvular interstitial cells on trilayered nanofibrous substrate mimicking morphologies of heart valve leaflet. Acta Biomater 2019; 85:142-156. [PMID: 30528607 PMCID: PMC6347416 DOI: 10.1016/j.actbio.2018.12.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/01/2018] [Accepted: 12/04/2018] [Indexed: 10/27/2022]
Abstract
Heart valve tissue engineering could be an alternative to the current bioprosthetic heart valve that faces limitations especially in pediatric patients. However, heart valve tissue engineering has remained challenging because leaflets - the primary component of a heart valve - have three layers with three diverse orientations - circumferential, random and radial, respectively. In order to mimic the orientations, we first designed three novel collectors to fabricate three nanofibrous layers with those orientations from a polymeric biomaterial in an electrospinning system. Then, we devised a novel direct electrospinning technique to develop a unified trilayered nanofibrous (TN) substrate comprising those oriented layers. The TN substrate supported the growth and orientations of seeded porcine valvular interstitial cells (PVICs) and their deposited collagen fibrils. After one month culture, the obtained trilayered tissue construct (TC) exhibited increased tensile properties over its TN substrate. Most importantly, the developed TC did not show any sign of shrinkage. Gene expression pattern of the PVICs indicated the developing stage of the TC. Their protein expression pattern was quite similar to that of leaflets. STATEMENT OF SIGNIFICANCE: This manuscript talks about development of a novel trilayered nanofibrous substrate mimicking the morphologies of a heart valve leaflet. It also describes culturing of valvular interstitial cells that reside in a leaflet, in the substrate and compares the behavior of the cultured cells with that in native leaflets in terms cell morphology, protein deposition and its orientation, and molecular signature. This study builds the groundwork for our future trilayered, tissue-engineered leaflet development. This research article would be of great interest to investigators and researchers in the field of cardiovascular tissue engineering especially in cardiac valve tissue engineering through biomaterial-based tissue engineering.
Collapse
Affiliation(s)
- Soumen Jana
- Division of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
25
|
Gomel MA, Lee R, Grande-Allen KJ. Comparing the Role of Mechanical Forces in Vascular and Valvular Calcification Progression. Front Cardiovasc Med 2019; 5:197. [PMID: 30687719 PMCID: PMC6335252 DOI: 10.3389/fcvm.2018.00197] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/20/2018] [Indexed: 01/07/2023] Open
Abstract
Calcification is a prevalent disease in most fully developed countries and is predominantly observed in heart valves and nearby vasculature. Calcification of either tissue leads to deterioration and, ultimately, failure causing poor quality of life and decreased overall life expectancy in patients. In valves, calcification presents as Calcific Aortic Valve Disease (CAVD), in which the aortic valve becomes stenotic when calcific nodules form within the leaflets. The initiation and progression of these calcific nodules is strongly influenced by the varied mechanical forces on the valve. In turn, the addition of calcific nodules creates localized disturbances in the tissue biomechanics, which affects extracellular matrix (ECM) production and cellular activation. In vasculature, atherosclerosis is the most common occurrence of calcification. Atherosclerosis exhibits as calcific plaque formation that forms in juxtaposition to areas of low blood shear stresses. Research in these two manifestations of calcification remain separated, although many similarities persist. Both diseases show that the endothelial layer and its regulation of nitric oxide is crucial to calcification progression. Further, there are similarities between vascular smooth muscle cells and valvular interstitial cells in terms of their roles in ECM overproduction. This review summarizes valvular and vascular tissue in terms of their basic anatomy, their cellular and ECM components and mechanical forces. Calcification is then examined in both tissues in terms of disease prediction, progression, and treatment. Highlighting the similarities and differences between these areas will help target further research toward disease treatment.
Collapse
|
26
|
Dekker S, van Geemen D, van den Bogaerdt AJ, Driessen-Mol A, Aikawa E, Smits AIPM. Sheep-Specific Immunohistochemical Panel for the Evaluation of Regenerative and Inflammatory Processes in Tissue-Engineered Heart Valves. Front Cardiovasc Med 2018; 5:105. [PMID: 30159315 PMCID: PMC6104173 DOI: 10.3389/fcvm.2018.00105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 07/13/2018] [Indexed: 12/27/2022] Open
Abstract
The creation of living heart valve replacements via tissue engineering is actively being pursued by many research groups. Numerous strategies have been described, aimed either at culturing autologous living valves in a bioreactor (in vitro) or inducing endogenous regeneration by the host via resorbable scaffolds (in situ). Whereas a lot of effort is being invested in the optimization of heart valve scaffold parameters and culturing conditions, the pathophysiological in vivo remodeling processes to which tissue-engineered heart valves are subjected upon implantation have been largely under-investigated. This is partly due to the unavailability of suitable immunohistochemical tools specific to sheep, which serves as the gold standard animal model in translational research on heart valve replacements. Therefore, the goal of this study was to comprise and validate a comprehensive sheep-specific panel of antibodies for the immunohistochemical analysis of tissue-engineered heart valve explants. For the selection of our panel we took inspiration from previous histopathological studies describing the morphology, extracellular matrix composition and cellular composition of native human heart valves throughout development and adult stages. Moreover, we included a range of immunological markers, which are particularly relevant to assess the host inflammatory response evoked by the implanted heart valve. The markers specifically identifying extracellular matrix components and cell phenotypes were tested on formalin-fixed paraffin-embedded sections of native sheep aortic valves. Markers for inflammation and apoptosis were tested on ovine spleen and kidney tissues. Taken together, this panel of antibodies could serve as a tool to study the spatiotemporal expression of proteins in remodeling tissue-engineered heart valves after implantation in a sheep model, thereby contributing to our understanding of the in vivo processes which ultimately determine long-term success or failure of tissue-engineered heart valves.
Collapse
Affiliation(s)
- Sylvia Dekker
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Daphne van Geemen
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | | | - Anita Driessen-Mol
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Anthal I. P. M. Smits
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
27
|
Nordquist E, LaHaye S, Nagel C, Lincoln J. Postnatal and Adult Aortic Heart Valves Have Distinctive Transcriptional Profiles Associated With Valve Tissue Growth and Maintenance Respectively. Front Cardiovasc Med 2018; 5:30. [PMID: 29740591 PMCID: PMC5928323 DOI: 10.3389/fcvm.2018.00030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/15/2018] [Indexed: 12/16/2022] Open
Abstract
Heart valves are organized connective tissues of high mechanical demand. They open and close over 100,000 times a day to preserve unidirectional blood flow by maintaining structure-function relationships throughout life. In affected individuals, structural failure compromises function and often leads to regurgitant blood flow and progressive heart failure. This is most common in degenerative valve disease due to age-related wear and tear, or congenital malformations. At present, the only effective treatment of valve disease is surgical repair or replacement and this is often impermanent and requires anti-coagulation therapy throughout life. Therefore, there is a critical need to discover new alternatives. A promising therapeutic area is tissue regeneration and in non-valvular tissues this requires a tightly regulated genetic “growth program” involving cell proliferation. To explore this in heart valves, we performed RNA-seq analysis to compare transcriptional profiles of aortic valve tissue isolated from mice during stages of growth (postnatal day (PND) 2) and adult maintenance (4 months). Data analysis reveals distinct mRNA profiles at each time point and pathway ontology identifies associated changes in biological functions. The PND2 aortic valve is characterized by extensive cell proliferation and expression of mRNAs related to the extracellular matrix (ECM). At 4 months, proliferation is not significant and a differential set of ECM-related genes are expressed. Interestingly there is enrichment of the defense response biological process at this later time point. Together, these data highlight the unique transcriptome of the postnatal valve during stages of growth and maturation, as well as biological functions associated with adult homeostatic valves. These studies create a platform for future work exploring the molecular programs altered in the onset of heart valve disease after birth and provide insights for the development of mechanistic-based therapies.
Collapse
Affiliation(s)
- Emily Nordquist
- Molecular Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, United States.,Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,The Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Stephanie LaHaye
- The Institute for Genomic Medicine at Nationwide Children's Hospital, Columbus, OH, United States
| | - Casey Nagel
- Ocean Ridge Biosciences, Deerfield Beach, FL, United States
| | - Joy Lincoln
- Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,The Heart Center, Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
28
|
Krishnamurthy VK, Stout AJ, Sapp MC, Matuska B, Lauer ME, Grande-Allen KJ. Dysregulation of hyaluronan homeostasis during aortic valve disease. Matrix Biol 2017; 62:40-57. [PMID: 27856308 PMCID: PMC10615645 DOI: 10.1016/j.matbio.2016.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 11/05/2016] [Accepted: 11/08/2016] [Indexed: 01/03/2023]
Abstract
Aortic valve disease (AVD) is one of the leading causes of cardiovascular mortality. Abnormal expression of hyaluronan (HA) and its synthesizing/degrading enzymes have been observed during latent AVD however, the mechanism of impaired HA homeostasis prior to and after the onset of AVD remains unexplored. Transforming growth factor beta (TGFβ) pathway defects and biomechanical dysfunction are hallmarks of AVD, however their association with altered HA regulation is understudied. Expression of HA homeostatic markers was evaluated in diseased human aortic valves and TGFβ1-cultured porcine aortic valve tissues using histology, immunohistochemistry and Western blotting. Further, porcine valve interstitial cell cultures were stretched (using Flexcell) and simultaneously treated with exogenous TGFβ1±inhibitors for activated Smad2/3 (SB431542) and ERK1/2 (U0126) pathways, and differential HA regulation was assessed using qRT-PCR. Pathological heavy chain HA together with abnormal regional expression of the enzymes HAS2, HYAL1, KIAA1199, TSG6 and IαI was demonstrated in calcified valve tissues identifying the collapse of HA homeostatic machinery during human AVD. Heightened TSG6 activity likely preceded the end-stage of disease, with the existence of a transitional, pre-calcific phase characterized by HA dysregulation. TGFβ1 elicited a fibrotic remodeling response in porcine aortic valves similar to human disease pathology, with increased collagen and HYAL to HAS ratio, and site-specific abnormalities in the expression of CD44 and RHAMM receptors. Further in these porcine valves, expression of HAS2 and HYAL1 was found to be differentially regulated by the Smad2/3 and ERK1/2 pathways, and CD44 expression was highly responsive to biomechanical strain. Leveraging the regulatory pathways that control both HA maintenance in normal valves and early postnatal dysregulation of HA homeostasis during disease may identify new mechanistic insight into AVD pathogenesis.
Collapse
Affiliation(s)
| | - Andrew J Stout
- Department of Materials Science and Nanoengineering, Rice University, Houston, TX 77005, USA
| | - Matthew C Sapp
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Brittany Matuska
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mark E Lauer
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
29
|
Angel PM, Baldwin HS, Gottlieb Sen D, Su YR, Mayer JE, Bichell D, Drake RR. Advances in MALDI imaging mass spectrometry of proteins in cardiac tissue, including the heart valve. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:927-935. [PMID: 28341601 DOI: 10.1016/j.bbapap.2017.03.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 02/15/2017] [Accepted: 03/20/2017] [Indexed: 01/01/2023]
Abstract
Significant progress has been made for tissue imaging of proteins using matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI IMS). These advancements now facilitate mapping of a wide range of proteins, peptides, and post-translational modifications in a wide variety of tissues; however, the use of MALDI IMS to detect proteins from cardiac tissue is limited. This review discusses the most recent advances in protein imaging and demonstrates application to cardiac tissue, including the heart valve. Protein imaging by MALDI IMS allows multiplexed histological mapping of proteins and protein components that are inaccessible by antibodies and should be considered an important tool for basic and clinical cardiovascular research. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.
Collapse
Affiliation(s)
- Peggi M Angel
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, USA; Medical University of South Carolina Proteomics Center, Medical University of South Carolina, Charleston, USA.
| | - H Scott Baldwin
- Department of Pediatrics and Cell Development and Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Yan Ru Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John E Mayer
- Department of Cardiac Surgery, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA
| | - David Bichell
- Division of Pediatric Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, USA; Medical University of South Carolina Proteomics Center, Medical University of South Carolina, Charleston, USA
| |
Collapse
|
30
|
Entstehung und Progression der Aortenklappendegeneration. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2017. [DOI: 10.1007/s00398-016-0086-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Assmann A, Struß M, Schiffer F, Heidelberg F, Munakata H, Timchenko EV, Timchenko PE, Kaufmann T, Huynh K, Sugimura Y, Leidl Q, Pinto A, Stoldt VR, Lichtenberg A, Akhyari P. Improvement of the in vivo
cellular repopulation of decellularized cardiovascular tissues by a detergent-free, non-proteolytic, actin-disassembling regimen. J Tissue Eng Regen Med 2017; 11:3530-3543. [DOI: 10.1002/term.2271] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 07/13/2016] [Accepted: 07/19/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Alexander Assmann
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Marc Struß
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Franziska Schiffer
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Friederike Heidelberg
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Hiroshi Munakata
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Elena V. Timchenko
- Department of Laser and Biotechnical Systems; Samara State Aerospace University; Samara Russia
| | - Pavel E. Timchenko
- Department of Laser and Biotechnical Systems; Samara State Aerospace University; Samara Russia
| | - Tim Kaufmann
- Department of Cardiovascular Engineering, Institute of Applied Medical Engineering, Helmholtz Institute; RWTH Aachen University; Aachen Germany
| | - Khon Huynh
- Institute of Haemostaseology, Haemotherapy and Transfusion Medicine; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Yukiharu Sugimura
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Quentin Leidl
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Antonio Pinto
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Volker R. Stoldt
- Institute of Haemostaseology, Haemotherapy and Transfusion Medicine; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Artur Lichtenberg
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| | - Payam Akhyari
- Department of Cardiovascular Surgery and Research Group for Experimental Surgery; Heinrich Heine University, Medical Faculty; Duesseldorf Germany
| |
Collapse
|
32
|
Zhang X, Xu B, Puperi DS, Wu Y, West JL, Grande-Allen KJ. Application of hydrogels in heart valve tissue engineering. J Long Term Eff Med Implants 2016; 25:105-34. [PMID: 25955010 DOI: 10.1615/jlongtermeffmedimplants.2015011817] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
With an increasing number of patients requiring valve replacements, there is heightened interest in advancing heart valve tissue engineering (HVTE) to provide solutions to the many limitations of current surgical treatments. A variety of materials have been developed as scaffolds for HVTE including natural polymers, synthetic polymers, and decellularized valvular matrices. Among them, biocompatible hydrogels are generating growing interest. Natural hydrogels, such as collagen and fibrin, generally show good bioactivity but poor mechanical durability. Synthetic hydrogels, on the other hand, have tunable mechanical properties; however, appropriate cell-matrix interactions are difficult to obtain. Moreover, hydrogels can be used as cell carriers when the cellular component is seeded into the polymer meshes or decellularized valve scaffolds. In this review, we discuss current research strategies for HVTE with an emphasis on hydrogel applications. The physicochemical properties and fabrication methods of these hydrogels, as well as their mechanical properties and bioactivities are described. Performance of some hydrogels including in vitro evaluation using bioreactors and in vivo tests in different animal models are also discussed. For future HVTE, it will be compelling to examine how hydrogels can be constructed from composite materials to replicate mechanical properties and mimic biological functions of the native heart valve.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning 110016, China
| | - Bin Xu
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Daniel S Puperi
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Yan Wu
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Jennifer L West
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | |
Collapse
|
33
|
Dybas J, Marzec KM, Pacia MZ, Kochan K, Czamara K, Chrabaszcz K, Staniszewska-Slezak E, Malek K, Baranska M, Kaczor A. Raman spectroscopy as a sensitive probe of soft tissue composition – Imaging of cross-sections of various organs vs. single spectra of tissue homogenates. Trends Analyt Chem 2016. [DOI: 10.1016/j.trac.2016.08.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Performance of allogeneic bioengineered replacement pulmonary valves in rapidly growing young lambs. J Thorac Cardiovasc Surg 2016; 152:1156-1165.e4. [DOI: 10.1016/j.jtcvs.2016.05.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/03/2016] [Accepted: 05/08/2016] [Indexed: 12/26/2022]
|
35
|
Ayoub S, Ferrari G, Gorman RC, Gorman JH, Schoen FJ, Sacks MS. Heart Valve Biomechanics and Underlying Mechanobiology. Compr Physiol 2016; 6:1743-1780. [PMID: 27783858 PMCID: PMC5537387 DOI: 10.1002/cphy.c150048] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
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.
Collapse
Affiliation(s)
- Salma Ayoub
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA
| | - Giovanni Ferrari
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Robert C. Gorman
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Joseph H. Gorman
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Frederick J. Schoen
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Michael S. Sacks
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA
| |
Collapse
|
36
|
van Geemen D, Soares ALF, Oomen PJA, Driessen-Mol A, Janssen-van den Broek MWJT, van den Bogaerdt AJ, Bogers AJJC, Goumans MJTH, Baaijens FPT, Bouten CVC. Age-Dependent Changes in Geometry, Tissue Composition and Mechanical Properties of Fetal to Adult Cryopreserved Human Heart Valves. PLoS One 2016; 11:e0149020. [PMID: 26867221 PMCID: PMC4750936 DOI: 10.1371/journal.pone.0149020] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/26/2016] [Indexed: 11/18/2022] Open
Abstract
There is limited information about age-specific structural and functional properties of human heart valves, while this information is key to the development and evaluation of living valve replacements for pediatric and adolescent patients. Here, we present an extended data set of structure-function properties of cryopreserved human pulmonary and aortic heart valves, providing age-specific information for living valve replacements. Tissue composition, morphology, mechanical properties, and maturation of leaflets from 16 pairs of structurally unaffected aortic and pulmonary valves of human donors (fetal-53 years) were analyzed. Interestingly, no major differences were observed between the aortic and pulmonary valves. Valve annulus and leaflet dimensions increase throughout life. The typical three-layered leaflet structure is present before birth, but becomes more distinct with age. After birth, cell numbers decrease rapidly, while remaining cells obtain a quiescent phenotype and reside in the ventricularis and spongiosa. With age and maturation-but more pronounced in aortic valves-the matrix shows an increasing amount of collagen and collagen cross-links and a reduction in glycosaminoglycans. These matrix changes correlate with increasing leaflet stiffness with age. Our data provide a new and comprehensive overview of the changes of structure-function properties of fetal to adult human semilunar heart valves that can be used to evaluate and optimize future therapies, such as tissue engineering of heart valves. Changing hemodynamic conditions with age can explain initial changes in matrix composition and consequent mechanical properties, but cannot explain the ongoing changes in valve dimensions and matrix composition at older age.
Collapse
Affiliation(s)
- Daphne van Geemen
- Soft Tissue Biomechanics & Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Ana L. F. Soares
- Soft Tissue Biomechanics & Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Pim J. A. Oomen
- Soft Tissue Biomechanics & Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Anita Driessen-Mol
- Soft Tissue Biomechanics & Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | | | - Antoon J. van den Bogaerdt
- Heart Valve Bank Rotterdam, Department of Cardio-Thoracic Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ad J. J. C. Bogers
- Heart Valve Bank Rotterdam, Department of Cardio-Thoracic Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Cardio-Thoracic Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Frank P. T. Baaijens
- Soft Tissue Biomechanics & Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Carlijn V. C. Bouten
- Soft Tissue Biomechanics & Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
37
|
Cells and extracellular matrix interplay in cardiac valve disease: because age matters. Basic Res Cardiol 2016; 111:16. [PMID: 26830603 DOI: 10.1007/s00395-016-0534-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/27/2015] [Accepted: 01/19/2016] [Indexed: 12/18/2022]
Abstract
Cardiovascular aging is a physiological process affecting all components of the heart. Despite the interest and experimental effort lavished on aging of cardiac cells, increasing evidence is pointing at the pivotal role of extracellular matrix (ECM) in cardiac aging. Structural and molecular changes in ECM composition during aging are at the root of significant functional modifications at the level of cardiac valve apparatus. Indeed, calcification or myxomatous degeneration of cardiac valves and their functional impairment can all be explained in light of age-related ECM alterations and the reciprocal interplay between altered ECM and cellular elements populating the leaflet, namely valvular interstitial cells and valvular endothelial cells, is additionally affecting valve function with striking reflexes on the clinical scenario. The initial experimental findings on this argument are underlining the need for a more comprehensive understanding on the biological mechanisms underlying ECM aging and remodeling as potentially constituting a pharmacological therapeutic target or a basis to improve existing prosthetic devices and treatment options. Given the lack of systematic knowledge on this topic, this review will focus on the ECM changes that occur during aging and on their clinical translational relevance and implications in the bedside scenario.
Collapse
|
38
|
Pierlot CM, Moeller AD, Lee JM, Wells SM. Pregnancy-induced remodeling of heart valves. Am J Physiol Heart Circ Physiol 2015; 309:H1565-78. [DOI: 10.1152/ajpheart.00816.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 09/06/2015] [Indexed: 01/13/2023]
Abstract
Recent studies have demonstrated remodeling of aortic and mitral valves leaflets under the volume loading and cardiac expansion of pregnancy. Those valves' leaflets enlarge with altered collagen fiber architecture, content, and cross-linking and biphasic changes (decreases, then increases) in extensibility during gestation. This study extends our analyses to right-sided valves, with additional compositional measurements for all valves. Valve leaflets were harvested from nonpregnant heifers and pregnant cows. Leaflet structure was characterized by leaflet dimensions, and ECM composition was determined using standard biochemical assays. Histological studies assessed changes in cellular and ECM components. Leaflet mechanical properties were assessed using equibiaxial mechanical testing. Collagen thermal stability and cross-linking were assessed using denaturation and hydrothermal isometric tension tests. Pulmonary and tricuspid leaflet areas increased during pregnancy by 35 and 55%, respectively. Leaflet thickness increased by 20% only in the pulmonary valve and largely in the fibrosa (30% thickening). Collagen crimp length was reduced in both the tricuspid (61%) and pulmonary (42%) valves, with loss of crimped area in the pulmonary valve. Thermomechanics showed decreased collagen thermal stability with surprisingly maintained cross-link maturity. The pulmonary leaflet exhibited the biphasic change in extensibility seen in left side valves, whereas the tricuspid leaflet mechanics remained largely unchanged throughout pregnancy. The tricuspid valve exhibits a remodeling response during pregnancy that is significantly diminished from the other three valves. All valves of the heart remodel in pregnancy in a manner distinct from cardiac pathology, with much similarity valve to valve, but with interesting valve-specific responses in the aortic and tricuspid valves.
Collapse
Affiliation(s)
- Caitlin M. Pierlot
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; and
| | - Andrew D. Moeller
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; and
| | - J. Michael Lee
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; and
- Department of Applied Oral Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sarah M. Wells
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; and
| |
Collapse
|
39
|
Levine RA, Hagége 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, Mérot J, Messas E, Milan DP, Neri T, Norris RA, Peal D, Perrocheau M, Probst V, Pucéat M, Rosenthal N, Solis J, Schott JJ, Schwammenthal E, Slaugenhaupt SA, Song JK, Yacoub MH. Mitral valve disease--morphology and mechanisms. Nat Rev Cardiol 2015; 12:689-710. [PMID: 26483167 DOI: 10.1038/nrcardio.2015.161] [Citation(s) in RCA: 260] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mitral valve disease is a frequent cause of heart failure and death. Emerging evidence indicates that the mitral valve is not a passive structure, but--even in adult life--remains dynamic and accessible for treatment. This concept motivates efforts to reduce the clinical progression of mitral valve disease through early detection and modification of underlying mechanisms. Discoveries of genetic mutations causing mitral valve elongation and prolapse have revealed that growth factor signalling and cell migration pathways are regulated by structural molecules in ways that can be modified to limit progression from developmental defects to valve degeneration with clinical complications. Mitral valve enlargement can determine left ventricular outflow tract obstruction in hypertrophic cardiomyopathy, and might be stimulated by potentially modifiable biological valvular-ventricular interactions. Mitral valve plasticity also allows adaptive growth in response to ventricular remodelling. However, adverse cellular and mechanobiological processes create relative leaflet deficiency in the ischaemic setting, leading to mitral regurgitation with increased heart failure and mortality. Our approach, which bridges clinicians and basic scientists, enables the correlation of observed disease with cellular and molecular mechanisms, leading to the discovery of new opportunities for improving the natural history of mitral valve disease.
Collapse
Affiliation(s)
- Robert A Levine
- Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Yawkey 5E, Boston, MA 02114, USA
| | - Albert A Hagége
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | | | | | - Jacob P Dal-Bianco
- Massachusetts General Hospital, Cardiac Ultrasound Laboratory, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Nabila Bouatia-Naji
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | - Patrick Bruneval
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | | | - Alain Carpentier
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | | | | | | | - Francesca N Delling
- Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, MA, USA
| | | | - Christian Dina
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | - Ronen Durst
- Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Leticia Fernandez-Friera
- Hospital Universitario HM Monteprincipe and the Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain
| | - Mark D Handschumacher
- Massachusetts General Hospital, Cardiac Ultrasound Laboratory, Harvard Medical School, Boston, MA, USA
| | | | - Xavier P Jeunemaitre
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | - Hervé Le Marec
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | - Thierry Le Tourneau
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | | | - Jean Mérot
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | - Emmanuel Messas
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | - David P Milan
- Cardiovascular Research Center, Harvard Medical School, Boston, MA, USA
| | - Tui Neri
- Aix-Marseille University, INSERM UMR 910, Marseille, France
| | | | - David Peal
- Cardiovascular Research Center, Harvard Medical School, Boston, MA, USA
| | - Maelle Perrocheau
- Hôpital Européen Georges Pompidou, Université René Descartes, UMR 970, Paris, France
| | - Vincent Probst
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | - Michael Pucéat
- Aix-Marseille University, INSERM UMR 910, Marseille, France
| | | | - Jorge Solis
- Hospital Universitario HM Monteprincipe and the Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain
| | - Jean-Jacques Schott
- University of Nantes, Thoracic Institute, INSERM UMR 1097, CNRS UMR 6291, Nantes, France
| | | | - Susan A Slaugenhaupt
- Center for Human Genetic Research, MGH Research Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
40
|
Guided tissue regeneration in heart valve replacement: from preclinical research to first-in-human trials. BIOMED RESEARCH INTERNATIONAL 2015; 2015:432901. [PMID: 26495295 PMCID: PMC4606187 DOI: 10.1155/2015/432901] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/21/2015] [Indexed: 11/18/2022]
Abstract
Heart valve tissue-guided regeneration aims to offer a functional and viable alternative to current prosthetic replacements. Not requiring previous cell seeding and conditioning in bioreactors, such exceptional tissue engineering approach is a very fascinating translational regenerative strategy. After in vivo implantation, decellularized heart valve scaffolds drive their same repopulation by recipient's cells for a prospective autologous-like tissue reconstruction, remodeling, and adaptation to the somatic growth of the patient. With such a viability, tissue-guided regenerated conduits can be delivered as off-the-shelf biodevices and possess all the potentialities for a long-lasting resolution of the dramatic inconvenience of heart valve diseases, both in children and in the elderly. A review on preclinical and clinical investigations of this therapeutic concept is provided with evaluation of the issues still to be well deliberated for an effective and safe in-human application.
Collapse
|
41
|
Duan B, Hockaday LA, Das S, Xu C, Butcher JT. Comparison of Mesenchymal Stem Cell Source Differentiation Toward Human Pediatric Aortic Valve Interstitial Cells within 3D Engineered Matrices. Tissue Eng Part C Methods 2015; 21:795-807. [PMID: 25594437 DOI: 10.1089/ten.tec.2014.0589] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Living tissue-engineered heart valves (TEHV) would be a major benefit for children who require a replacement with the capacity for growth and biological integration. A persistent challenge for TEHV is accessible human cell source(s) that can mimic native valve cell phenotypes and matrix remodeling characteristics that are essential for long-term function. Mesenchymal stem cells derived from bone marrow (BMMSC) or adipose tissue (ADMSC) are intriguing cell sources for TEHV, but they have not been compared with pediatric human aortic valve interstitial cells (pHAVIC) in relevant 3D environments. In this study, we compared the spontaneous and induced multipotency of ADMSC and BMMSC with that of pHAVIC using different induction media within three-dimensional (3D) bioactive hybrid hydrogels with material modulus comparable to that of aortic heart valve leaflets. pHAVIC possessed some multi-lineage differentiation capacity in response to induction media, but limited to the earliest stages and much less potent than either ADMSC or BMMSC. ADMSC expressed cell phenotype markers more similar to pHAVIC when conditioned in basic fibroblast growth factor (bFGF) containing HAVIC growth medium, while BMMSC generally expressed similar extracellular matrix remodeling characteristics to pHAVIC. Finally, we covalently attached bFGF to PEG monoacrylate linkers and further covalently immobilized in the 3D hybrid hydrogels. Immobilized bFGF upregulated vimentin expression and promoted the fibroblastic differentiation of pHAVIC, ADMSC, and BMMSC. These findings suggest that stem cells retain a heightened capacity for osteogenic differentiation in 3D culture, but can be shifted toward fibroblast differentiation through matrix tethering of bFGF. Such a strategy is likely important for utilizing stem cell sources in heart valve tissue engineering applications.
Collapse
Affiliation(s)
- Bin Duan
- 1 Department of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Laura A Hockaday
- 1 Department of Biomedical Engineering, Cornell University , Ithaca, New York
| | - Shoshana Das
- 2 Department of Biological and Environmental Engineering, Cornell University , Ithaca, New York
| | - Charlie Xu
- 2 Department of Biological and Environmental Engineering, Cornell University , Ithaca, New York
| | - Jonathan T Butcher
- 1 Department of Biomedical Engineering, Cornell University , Ithaca, New York
| |
Collapse
|
42
|
Czamara K, Natorska J, Kapusta P, Baranska M, Kaczor A. Raman microspectroscopy of human aortic valves: investigation of the local and global biochemical changes associated with calcification in aortic stenosis. Analyst 2015; 140:2164-70. [DOI: 10.1039/c4an01856g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Raman microspectroscopy was applied to characterize the local and global biochemical changes associated with calcification in human stenotic aortic valves.
Collapse
Affiliation(s)
- Krzysztof Czamara
- Faculty of Chemistry
- Jagiellonian University
- 30-060 Krakow
- Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET)
| | - Joanna Natorska
- Institute of Cardiology
- Jagiellonian University
- 31-202 Krakow
- Poland
| | | | - Malgorzata Baranska
- Faculty of Chemistry
- Jagiellonian University
- 30-060 Krakow
- Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET)
| | - Agnieszka Kaczor
- Faculty of Chemistry
- Jagiellonian University
- 30-060 Krakow
- Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET)
| |
Collapse
|
43
|
Duan B, Kapetanovic E, Hockaday LA, Butcher JT. Three-dimensional printed trileaflet valve conduits using biological hydrogels and human valve interstitial cells. Acta Biomater 2014; 10:1836-46. [PMID: 24334142 DOI: 10.1016/j.actbio.2013.12.005] [Citation(s) in RCA: 253] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 11/20/2013] [Accepted: 12/05/2013] [Indexed: 12/20/2022]
Abstract
Tissue engineering has great potential to provide a functional de novo living valve replacement, capable of integration with host tissue and growth. Among various valve conduit fabrication techniques, three-dimensional (3-D) bioprinting enables deposition of cells and hydrogels into 3-D constructs with anatomical geometry and heterogeneous mechanical properties. Successful translation of this approach, however, is constrained by the dearth of printable and biocompatible hydrogel materials. Furthermore, it is not known how human valve cells respond to these printed environments. In this study, 3-D printable formulations of hybrid hydrogels are developed, based on methacrylated hyaluronic acid (Me-HA) and methacrylated gelatin (Me-Gel), and used to bioprint heart valve conduits containing encapsulated human aortic valvular interstitial cells (HAVIC). Increasing Me-Gel concentration resulted in lower stiffness and higher viscosity, facilitated cell spreading, and better maintained HAVIC fibroblastic phenotype. Bioprinting accuracy was dependent upon the relative concentrations of Me-Gel and Me-HA, but when optimized enabled the fabrication of a trileaflet valve shape accurate to the original design. HAVIC encapsulated within bioprinted heart valves maintained high viability, and remodeled the initial matrix by depositing collagen and glyosaminoglycans. These findings represent the first rational design of bioprinted trileaflet valve hydrogels that regulate encapsulated human VIC behavior. The use of anatomically accurate living valve scaffolds through bioprinting may accelerate understanding of physiological valve cell interactions and progress towards de novo living valve replacements.
Collapse
Affiliation(s)
- B Duan
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - E Kapetanovic
- College of Human Ecology, Cornell University, Ithaca, NY, USA
| | - L A Hockaday
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - J T Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
44
|
Abstract
Calcific aortic valve disease (CAVD) is a major contributor to cardiovascular morbidity and mortality and, given its association with age, the prevalence of CAVD is expected to continue to rise as global life expectancy increases. No drug strategies currently exist to prevent or treat CAVD. Given that valve replacement is the only available clinical option, patients often cope with a deteriorating quality of life until diminished valve function demands intervention. The recognition that CAVD results from active cellular mechanisms suggests that the underlying pathways might be targeted to treat the condition. However, no such therapeutic strategy has been successfully developed to date. One hope was that drugs already used to treat vascular complications might also improve CAVD outcomes, but the mechanisms of CAVD progression and the desired therapeutic outcomes are often different from those of vascular diseases. Therefore, we discuss the benchmarks that must be met by a CAVD treatment approach, and highlight advances in the understanding of CAVD mechanisms to identify potential novel therapeutic targets.
Collapse
Affiliation(s)
- Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, 3 Blackfan Circle, 17th Floor, Center for Life Sciences Boston, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, 3 Blackfan Circle, 17th Floor, Center for Life Sciences Boston, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - W David Merryman
- Department of Biomedical Engineering, 2213 Garland Avenue, Vanderbilt University, Nashville, TN 37212, USA
| |
Collapse
|
45
|
Buchanan RM, Sacks MS. Interlayer micromechanics of the aortic heart valve leaflet. Biomech Model Mechanobiol 2013; 13:813-26. [PMID: 24292631 DOI: 10.1007/s10237-013-0536-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 10/09/2013] [Indexed: 10/26/2022]
Abstract
While the mechanical behaviors of the fibrosa and ventricularis layers of the aortic valve (AV) leaflet are understood, little information exists on their mechanical interactions mediated by the GAG-rich central spongiosa layer. Parametric simulations of the interlayer interactions of the AV leaflets in flexure utilized a tri-layered finite element (FE) model of circumferentially oriented tissue sections to investigate inter-layer sliding hypothesized to occur. Simulation results indicated that the leaflet tissue functions as a tightly bonded structure when the spongiosa effective modulus was at least 25 % that of the fibrosa and ventricularis layers. Novel studies that directly measured transmural strain in flexure of AV leaflet tissue specimens validated these findings. Interestingly, a smooth transmural strain distribution indicated that the layers of the leaflet indeed act as a bonded unit, consistent with our previous observations (Stella and Sacks in J Biomech Eng 129:757-766, 2007) of a large number of transverse collagen fibers interconnecting the fibrosa and ventricularis layers. Additionally, when the tri-layered FE model was refined to match the transmural deformations, a layer-specific bimodular material model (resulting in four total moduli) accurately matched the transmural strain and moment-curvature relations simultaneously. Collectively, these results provide evidence, contrary to previous assumptions, that the valve layers function as a bonded structure in the low-strain flexure deformation mode. Most likely, this results directly from the transverse collagen fibers that bind the layers together to disable physical sliding and maintain layer residual stresses. Further, the spongiosa may function as a general dampening layer while the AV leaflets deforms as a homogenous structure despite its heterogeneous architecture.
Collapse
Affiliation(s)
- Rachel M Buchanan
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA
| | | |
Collapse
|
46
|
Balaoing LR, Post AD, Liu H, Minn KT, Grande-Allen KJ. Age-related changes in aortic valve hemostatic protein regulation. Arterioscler Thromb Vasc Biol 2013; 34:72-80. [PMID: 24177329 DOI: 10.1161/atvbaha.113.301936] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although valvular endothelial cells have unique responses compared with vascular endothelial cells, valvular regulation of hemostasis is not well-understood. Heart valves remodel throughout a person's lifetime, resulting in changes in extracellular matrix composition and tissue mechanical properties that may affect valvular endothelial cell hemostatic function. This work assessed valvular endothelial cell regulation of hemostasis in situ and in vitro as a function of specimen age. APPROACH AND RESULTS Porcine aortic valves were assigned to 1 of 3 age groups: Young (YNG) (6 weeks); Adult (ADT) (6 months); or Elderly (OLD) (2 years). Histological examination of valves showed that secreted thrombotic/antithrombotic proteins localize at the valve endothelium and tissue interior. Gene expression and immunostains for von Willebrand factor (VWF), tissue factor pathway inhibitor, and tissue plasminogen activator in YNG porcine aortic valve endothelial cells were higher than they were for OLD, whereas plasminogen activator inhibitor 1 levels in OLD were higher than those for YNG and ADT. Histamine-stimulated YNG porcine aortic valve endothelial cells released higher concentrations of VWF proteins than OLD, and the fractions of VWF-140 fragments was not different between age groups. A calcific aortic valve disease in vitro model using valvular interstitial cells confirmed that VWF in culture significantly increased valvular interstitial cell nodule formation and calcification. CONCLUSIONS Hemostatic protein regulation in aortic valve tissues and in valvular endothelial cells changes with age. The presence of VWF and other potential hemostatic proteins increase valvular interstitial cell calcification in vitro. Therefore, the increased capacity of elderly valves to sequester the hemostatic proteins, together with age-associated loss of extracellular matrix organization, warrants investigation into potential role of these proteins in the formation of calcific nodules.
Collapse
Affiliation(s)
- Liezl R Balaoing
- From the Department of Bioengineering, Rice University, Houston, TX
| | | | | | | | | |
Collapse
|
47
|
Abstract
The aortic valve is highly responsive to cyclical and continuous mechanical forces, at the macroscopic and cellular levels. In this report, we delineate mechanokinetics (effects of mechanical inputs on the cells) and mechanodynamics (effects of cells and pathologic processes on the mechanics) of the aortic valve, with a particular focus on how mechanical inputs synergize with the inflammatory cytokine and other biomolecular signaling to contribute to the process of aortic valve calcification.
Collapse
|
48
|
Pham T, Sun W. Material properties of aged human mitral valve leaflets. J Biomed Mater Res A 2013; 102:2692-703. [PMID: 24039052 DOI: 10.1002/jbm.a.34939] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 08/01/2013] [Accepted: 08/26/2013] [Indexed: 11/08/2022]
Abstract
This study aimed to characterize the mechanical properties of aged human anterior mitral leaflets (AML) and posterior mitral leaflets (PML). The AML and PML samples from explanted human hearts (n = 21, mean age of 82.62 ± 8.77-years-old) were subjected to planar biaxial mechanical tests. The material stiffness, extensibility, and degree of anisotropy of the leaflet samples were quantified. The microstructure of the samples was assessed through histology. Both the AML and PML samples exhibited a nonlinear and anisotropic behavior with the circumferential direction being stiffer than the radial direction. The AML samples were significantly stiffer than the PML samples in both directions, suggesting that they should be modeled with separate sets of material properties in computational studies. Histological analysis indicated the changes in the tissue elastic constituents, including the fragmented and disorganized elastin network, the presence of fibrosis and proteoglycan/glycosaminoglycan infiltration and calcification, suggesting possible valvular degenerative characteristics in the aged human leaflet samples. Overall, stiffness increased and areal strain decreased with calcification severity. In addition, leaflet tissues from hypertensive individuals also exhibited a higher stiffness and low areal strain than normotensive individuals. There are significant differences in the mechanical properties of the two human mitral valve leaflets from this advanced age group. The morphologic changes in the tissue composition and structure also infer the structural and functional difference between aged human valves and those of animals.
Collapse
Affiliation(s)
- Thuy Pham
- Tissue Mechanics Laboratory, Department of Mechanical Engineering, Biomedical Engineering Program, University of Connecticut, Storrs, Connecticut, 06269
| | | |
Collapse
|
49
|
Glycosaminoglycan entrapment by fibrin in engineered heart valve tissues. Acta Biomater 2013; 9:8149-57. [PMID: 23791855 DOI: 10.1016/j.actbio.2013.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 05/27/2013] [Accepted: 06/06/2013] [Indexed: 02/05/2023]
Abstract
Tissue engineered heart valves (TEHVs) may provide a permanent solution to congenital heart valve disease by permitting somatic valve growth in the pediatric patient. However, to date, TEHV studies have focused primarily on collagen, the dominant component of valve extracellular matrix (ECM). Temporal decreases in other ECM components, such as the glycosaminoglycans (GAGs), generally decrease as cells produce more collagen under mechanically loaded states; nevertheless, GAGs represent a key component of the valve ECM, providing structural stability and hydration to the leaflets. In an effort to retain GAGs within the engineered constructs, here we investigated the utility of the protein fibrin in combination with a valve-like, cyclic flexure and steady flow (flex-flow) mechanical conditioning culture process using adult human periodontal ligament cells (PLCs). We found both fibrin and flex-flow mechanical components to be independently significant (p<0.05), and hence important in influencing the DNA, GAG and collagen contents of the engineered tissues. In addition, the interaction of fibrin with flex-flow was found to be significant in the case of collagen; specifically, the combination of these environments promoted PLC collagen production resulting in a significant difference compared to dynamic and statically cultured specimens without fibrin. Histological examination revealed that the GAGs were retained by fibrin entrapment and adhesion, which were subsequently confirmed by additional experiments on native valve tissues. We conclude that fibrin in the flex-flow culture of engineered heart valve tissues: (i) augments PLC-derived collagen production; and (ii) enhances retention of GAGs within the developing ECM.
Collapse
|
50
|
Bloch O, Erdbrügger W, Völker W, Schenk A, Posner S, Konertz W, Dohmen PM. Extracellular matrix in deoxycholic acid decellularized aortic heart valves. Med Sci Monit 2013. [PMID: 23207452 PMCID: PMC3560792 DOI: 10.12659/msm.883618] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Only limited information is available regarding the influence of decellularization on the extracellular matrix in heart valves. Within the extracellular matrix proteoglycans (PG) play a central role in the structural organization and physical functioning of valves and in their capability of settling with endothelial and interstitial cells partially myofibroblasts. We have therefore estimated the effects of decellularization using deoxycholic acid on the structure of the extracellular matrix and PG’s in porcine aortic valves. Material/Methods Cupromeronic blue was used, alone or in combination with OsO4/thio-carbo-hydrazide/OsO4 for electron microscopic visualization. For PG and glycosaminoglycan (GAG) investigation a papain digestion was employed in combination with photometric determination using dimethylmethylene blue. Results The results indicate that deoxycholic acid affects the compartmentation of the PG-associated interstitial network not significantly. Compared to controls the PG-rich network was preserved even after deoxycholic acid treatment for 48 h. In parallel to electron microscopy immune assays (ELISA) showed smooth muscle cell α-actin to be reduced to 0.96%±0.71 and total soluble protein to 6.68%±2.0 (n=3) of untreated controls. Protein loss corresponded well with the observations in electron micrographs of rupture and efflux of cell content. Further signs of lysis were irregular cell contours and loss of the basement membrane. Conclusions Efficient cell-lysis without disintegration or loss of integrity of the interstitial PG network can be achieved by treatment of aortic valves with deoxycholic acid for 48h. This protocol might also be suitable for clinical use to optimize conditions for growth and autologous remodelling of valves.
Collapse
Affiliation(s)
- Oliver Bloch
- Department of Cardiovascular Surgery, Charité, Medical University Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|