1
|
He Y, Yang YJ, Wang ZJ, Tang L. Bibliometric analysis of treatment modalities in calcific aortic valve stenosis. Front Pharmacol 2025; 16:1431311. [PMID: 40183078 PMCID: PMC11966050 DOI: 10.3389/fphar.2025.1431311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
Background Calcific aortic valve stenosis (CAVS) is a common cardiovascular condition associated with significant adverse events and high mortality rates. Unfortunately, there are currently no effective pharmacological treatments to halt or prevent its progression. Through our analysis of global trends and treatment strategies, we have identified valuable insights and promising therapeutic possibilities. Additionally, by utilizing bibliometric and visualization techniques, we provide a comprehensive overview of the current research landscape in this field. Method According to our design idea, we used the Web of Science database to select publications on aortic stenosis and related treatments. Through our VOSviewer and CiteSpace analysis, a total of 787 articles have been analyzed by September 2024. We also summarize and explore the most prolific authors, the most prolific countries, and the journals and institutions that publish the most articles. Results A visual analysis of the collected articles reveals that Canada and the United States have the highest publication volumes in this field. Among institutions, Harvard University in the U.S. leads in publication count, followed by Laval University in Canada and the University of California in the U.S. The top three research hotspots are stenosis, calcification, and progression. The journal with the highest number of publications in this area is Frontiers in Cardiovascular Medicine, followed by Catheterization and Cardiovascular Interventions and Arteriosclerosis, Thrombosis and Vascular Biology. Furthermore, research on CAVS treatment spans various directions and focuses, including therapeutic approaches, pathogenesis, and diagnostic methods. Conclusion Research into CAVS treatment has advanced significantly over the years. While interventional and surgical valve replacement remains the mainstay treatments for aortic stenosis, they are insufficient to fully meet the needs of the patient. Emerging priorities now focus on improving diagnostics, exploring innovative therapies, uncovering disease mechanisms, and developing novel drugs. These findings highlight the evolving demands in this field and underscore the need for continued research to address these challenges.
Collapse
Affiliation(s)
| | | | | | - Liang Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Prabhakar AP, Lopez-Candales A. Calcific aortic valve disease and cardiometabolic triggers: an explanation behind progression of aortic valvular disease and failure of medical therapy interventions. Postgrad Med 2024; 136:810-818. [PMID: 39297302 DOI: 10.1080/00325481.2024.2406740] [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: 02/20/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
Calcific aortic valve disease (CAVD), a nonrheumatic stenosis of the trileaflet aortic valve, is a complex, multifaceted cardiovascular condition involving a widespread inflammatory process and an analogous atheromatous process affecting the arteries. It is currently the most encountered valvular abnormality in cardiology. Although distinctive abnormal mechanical forces are at the core propelling a responsive mechanosensitive feedback cascade, implicated in both initiation and perpetuation of CAVD; we propose a conundrum of metabolic abnormalities including hypertension, elevated fasting blood sugar, decreased high-density lipoprotein, hypertriglyceridemia, and abdominal obesity as perpetuators of this process. Furthermore, we suggest CAVD as a cardio metabolic disorder. New perspectives as well as which pathways we believe are critically involved and ideas for early intervention are discussed.
Collapse
Affiliation(s)
- Akruti Patel Prabhakar
- Advanced Cardiac Imaging Research Fellow, Department of Cardiovascular Medicine, University of Louisville, Louisville, KY, USA
| | - Angel Lopez-Candales
- Cardiology Section, Dayton VA Medical Center, Wright State Boonshoft School of Medicine, Dayton, OH, USA
| |
Collapse
|
3
|
Xie M, Cao H, Qiao W, Yan G, Qian X, Zhang Y, Xu L, Wen S, Shi J, Cheng M, Dong N. Shear stress activates the Piezo1 channel to facilitate valvular endothelium-oriented differentiation and maturation of human induced pluripotent stem cells. Acta Biomater 2024; 178:181-195. [PMID: 38447808 DOI: 10.1016/j.actbio.2024.02.043] [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: 07/27/2023] [Revised: 02/15/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Valvular endothelial cells (VECs) derived from human induced pluripotent stem cells (hiPSCs) provide an unlimited cell source for tissue engineering heart valves (TEHVs); however, they are limited by their low differentiation efficiency and immature function. In our study, we applied unidirectional shear stress to promote hiPSCs differentiation into valvular endothelial-like cells (VELs). Compared to the static group, shear stress efficiently promoted the differentiation and functional maturation of hiPSC-VELs, as demonstrated by the efficiency of endothelial differentiation reaching 98.3% in the high shear stress group (45 dyn/cm2). Furthermore, we found that Piezo1 served as a crucial mechanosensor for the differentiation and maturation of VELs. Mechanistically, the activation of Piezo1 by shear stress resulted in the influx of calcium ions, which in turn initiated the Akt signaling pathway and promoted the differentiation of hiPSCs into mature VELs. Moreover, VELs cultured on decellularized heart valves (DHVs) exhibited a notable propensity for proliferation, robust adhesion properties, and antithrombotic characteristics, which were dependent on the activation of the Piezo1 channel. Overall, our study demonstrated that proper shear stress activated the Piezo1 channel to facilitate the differentiation and maturation of hiPSC-VELs via the Akt pathway, providing a potential cell source for regenerative medicine, drug screening, pathogenesis, and disease modeling. STATEMENT OF SIGNIFICANCE: This is the first research that systematically analyzes the effect of shear stress on valvular endothelial-like cells (VELs) derived from human induced pluripotent stem cells (hiPSCs). Mechanistically, unidirectional shear stress activates Piezo1, resulting in an elevation of calcium levels, which triggers the Akt signaling pathway and then facilitates the differentiation of functional maturation VELs. After exposure to shear stress, the VELs exhibited enhanced proliferation, robust adhesion capabilities, and antithrombotic characteristics while being cultured on decellularized heart valves. Thus, it is of interest to develop hiPSCs-VELs using shear stress and the Piezo1 channel provides insights into the functional maturation of valvular endothelial cells, thereby serving as a catalyst for potential applications in the development of therapeutic and tissue-engineered heart valves in the future.
Collapse
Affiliation(s)
- Minghui Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hong Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ge Yan
- Department of Cardiovascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Xingyu Qian
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuyu Wen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
4
|
Yen Y, Hung KC, Chan YH, Wu VCC, Cheng YT, Lin CP, Yeh JK, Chu PH, Chen SW. Association of Valve Size and Hemodynamic Performance With Clinical Outcomes in Aortic Valve Replacement - A Long-Term Follow-up in an Asian Population. Circ J 2024; 88:559-567. [PMID: 37019644 DOI: 10.1253/circj.cj-22-0718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
BACKGROUND Studies of the influence of smaller body type on the severity of prosthesis-patient mismatch (PPM) after small-sized surgical aortic valve replacement (SAVR) are few, but the issue is particularly relevant for Asian patients. METHODS AND RESULTS 695 patients who underwent SAVR with bioprosthetic valves had their hemodynamic valve performance analyzed at 3 months, 1 year, 3 years, and 5 years after operation, and clinical outcomes were assessed. The patients were stratified into 3 valve size groups: 19/21, 23, and 25/27 mm. A smaller valve was associated with higher mean pressure gradients at the 4 time points after operation (P trend <0.05). However, the 3 valve size groups demonstrated no significant differences in the risk of clinical events. At none of the time points did patients with projected PPM show increased mean pressure gradients (P>0.05), whereas patients with measured PPM did (P<0.05). Compared with patients with projected PPM, those with measured PPM demonstrated higher rates of infective endocarditis readmission (adjusted hazard ratio [aHR] 3.31, 95% confidence interval [CI] 1.06-10.39) and a higher risk of composite outcomes (aHR 1.45, 95% CI 0.95-2.22, P=0.087). CONCLUSIONS Relative to those receiving larger valves, patients receiving small bioprosthetic valves had poorer hemodynamic performance but did not demonstrate differences in clinical events in long-term follow-up.
Collapse
Affiliation(s)
- Yi Yen
- Department of Medical Education, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung Memorial Hospital, Chiayi, Chang Gung University
| | - Kuo-Chun Hung
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University
| | - Yi-Hsin Chan
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University
| | - Victor Chien-Chia Wu
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University
| | - Yu-Ting Cheng
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University
| | - Chia-Pin Lin
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University
| | - Jih-Kai Yeh
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University
| | - Pao-Hsien Chu
- Department of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University
| | - Shao-Wei Chen
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Linkou Medical Center
| |
Collapse
|
5
|
Jiang H, Liu M, Qin Y, Zhang H. miR-9 promotes canine endothelial-like cell migration by targeting COL15A1. Vet Med Sci 2024; 10:e1339. [PMID: 38109263 PMCID: PMC10766037 DOI: 10.1002/vms3.1339] [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: 07/15/2023] [Revised: 11/16/2023] [Accepted: 12/03/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Endothelial cell migration is the initial stage of angiogenesis. In previous studies, miR-9 has been found to regulate angiogenesis and cell migration in human medicine. OBJECTIVES This study aimed to reveal the regulatory effect of miR-9 on canine endothelial cell migration. METHODS Embryonic canine ventricle myocardium tissues were collected and induced to differentiate into endothelial-like cells (ELCs). A transwell and invasion assay were used to evaluate the impact of miR-9 on the migration capacity of ELCs, after which a luciferase reporter assay, western blotting, RNA sequencing and reverse transcription-polymerase chain reaction were conducted to explore the regulatory mechanism. RESULTS Our results showed that we successfully induced the primary cells derived from canine cardiac embryo tissues into ELCs. MiR-9 also promoted the migration and invasion of canine ELCs, and inhibited the expression of collagen XV, an angiogenic inhibitor, at the translational level by targeting the 3' untranslated region of COL15A1 gene. Furthermore, RNA sequencing showed that overexpression of miR-9 impacted several signalling pathways and eight genes involved in angiogenesis and cell migration in canine ELCs. CONCLUSIONS These findings suggest that miR-9 enhances the migration of canine ELCs and may serve as a potential diagnostic and therapeutic target for canine diseases involved in endothelial cells migration and angiogenesis, but more further studies are needed.
Collapse
Affiliation(s)
- Heng Jiang
- Institute of Tropical Agriculture and ForestryHainan UniversityHainanPR China
| | - Mengmeng Liu
- Institute of Tropical Agriculture and ForestryHainan UniversityHainanPR China
- One Health InstituteHainan UniversityHainanPR China
| | - Yao Qin
- Institute of Tropical Agriculture and ForestryHainan UniversityHainanPR China
| | - Hong Zhang
- Institute of Tropical Agriculture and ForestryHainan UniversityHainanPR China
| |
Collapse
|
6
|
Ibrahim DM, Fomina A, Bouten CVC, Smits AIPM. Functional regeneration at the blood-biomaterial interface. Adv Drug Deliv Rev 2023; 201:115085. [PMID: 37690484 DOI: 10.1016/j.addr.2023.115085] [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: 10/31/2022] [Revised: 06/01/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
The use of cardiovascular implants is commonplace in clinical practice. However, reproducing the key bioactive and adaptive properties of native cardiovascular tissues with an artificial replacement is highly challenging. Exciting new treatment strategies are under development to regenerate (parts of) cardiovascular tissues directly in situ using immunomodulatory biomaterials. Direct exposure to the bloodstream and hemodynamic loads is a particular challenge, given the risk of thrombosis and adverse remodeling that it brings. However, the blood is also a source of (immune) cells and proteins that dominantly contribute to functional tissue regeneration. This review explores the potential of the blood as a source for the complete or partial in situ regeneration of cardiovascular tissues, with a particular focus on the endothelium, being the natural blood-tissue barrier. We pinpoint the current scientific challenges to enable rational engineering and testing of blood-contacting implants to leverage the regenerative potential of the blood.
Collapse
Affiliation(s)
- Dina M Ibrahim
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Aleksandra Fomina
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Graduate School of Life Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Anthal I P M Smits
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
7
|
Shu L, Yuan Z, Li F, Cai Z. Oxidative stress and valvular endothelial cells in aortic valve calcification. Biomed Pharmacother 2023; 163:114775. [PMID: 37116353 DOI: 10.1016/j.biopha.2023.114775] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/15/2023] [Accepted: 04/23/2023] [Indexed: 04/30/2023] Open
Abstract
Calcified aortic valve disease (CAVD) is a common cardiovascular disease in elderly individuals. Although it was previously considered a degenerative disease, it is, in fact, a progressive disease involving multiple mechanisms. Aortic valve endothelial cells, which cover the outermost layer of the aortic valve and are directly exposed to various pathogenic factors, play a significant role in the onset and progression of CAVD. Hemodynamic changes can directly damage the structure and function of valvular endothelial cells (VECs). This leads to inflammatory infiltration and oxidative stress, which promote the progression of CAVD. VECs can regulate the pathological differentiation of valvular interstitial cells (VICs) through NO and thus affect the process of CAVD. Under the influence of pathological factors, VECs can also be transformed into VICs through EndMT, and then the pathological differentiation of VICs eventually leads to the formation of calcification. This review discusses the role of VECs, especially the role of oxidative stress in VECs, in the process of aortic valve calcification.
Collapse
Affiliation(s)
- Li Shu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Zhen Yuan
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan 430022, Hubei, China.
| | - Zhejun Cai
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China.
| |
Collapse
|
8
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
9
|
Lim S, Kim SW, Kim IK, Song BW, Lee S. Organ-on-a-chip: Its use in cardiovascular research. Clin Hemorheol Microcirc 2023; 83:315-339. [PMID: 36502306 DOI: 10.3233/ch-221428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Organ-on-a-chip (OOAC) has attracted great attention during the last decade as a revolutionary alternative to conventional animal models. This cutting-edge technology has also brought constructive changes to the field of cardiovascular research. The cardiovascular system, especially the heart as a well-protected vital organ, is virtually impossible to replicate in vitro with conventional approaches. This made scientists assume that they needed to use animal models for cardiovascular research. However, the frequent failure of animal models to correctly reflect the native cardiovascular system necessitated a search for alternative platforms for preclinical studies. Hence, as a promising alternative to conventional animal models, OOAC technology is being actively developed and tested in a wide range of biomedical fields, including cardiovascular research. Therefore, in this review, the current literature on the use of OOACs for cardiovascular research is presented with a focus on the basis for using OOACs, and what has been specifically achieved by using OOACs is also discussed. By providing an overview of the current status of OOACs in cardiovascular research and its future perspectives, we hope that this review can help to develop better and optimized research strategies for cardiovascular diseases (CVDs) as well as identify novel applications of OOACs in the near future.
Collapse
Affiliation(s)
- Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| |
Collapse
|
10
|
Majumdar U, Choudhury TZ, Manivannan S, Ueyama Y, Basu M, Garg V. Single-cell RNA-sequencing analysis of aortic valve interstitial cells demonstrates the regulation of integrin signaling by nitric oxide. Front Cardiovasc Med 2022; 9:742850. [PMID: 36386365 PMCID: PMC9640371 DOI: 10.3389/fcvm.2022.742850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is an increasingly prevalent condition among the elderly population that is associated with significant morbidity and mortality. Insufficient understanding of the underlying disease mechanisms has hindered the development of pharmacologic therapies for CAVD. Recently, we described nitric oxide (NO) mediated S-nitrosylation as a novel mechanism for preventing the calcific process. We demonstrated that NO donor or an S-nitrosylating agent, S-nitrosoglutathione (GSNO), inhibits spontaneous calcification in porcine aortic valve interstitial cells (pAVICs) and this was supported by single-cell RNA sequencing (scRNAseq) that demonstrated NO donor and GSNO inhibited myofibroblast activation of pAVICs. Here, we investigated novel signaling pathways that are critical for the calcification of pAVICs that are altered by NO and GSNO by performing an in-depth analysis of the scRNA-seq dataset. Transcriptomic analysis revealed 1,247 differentially expressed genes in pAVICs after NO donor or GSNO treatment compared to untreated cells. Pathway-based analysis of the differentially expressed genes revealed an overrepresentation of the integrin signaling pathway, along with the Rho GTPase, Wnt, TGF-β, and p53 signaling pathways. We demonstrate that ITGA8 and VCL, two of the identified genes from the integrin signaling pathway, which are known to regulate cell-extracellular matrix (ECM) communication and focal adhesion, were upregulated in both in vitro and in vivo calcific conditions. Reduced expression of these genes after treatment with NO donor suggests that NO inhibits calcification by targeting myofibroblast adhesion and ECM remodeling. In addition, withdrawal of NO donor after 3 days of exposure revealed that NO-mediated transcriptional and translational regulation is a transient event and requires continuous NO exposure to inhibit calcification. Overall, our data suggest that NO and S-nitrosylation regulate the integrin signaling pathway to maintain healthy cell-ECM interaction and prevent CAVD.
Collapse
Affiliation(s)
- Uddalak Majumdar
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Talita Z. Choudhury
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Sathiyanarayanan Manivannan
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Yukie Ueyama
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Madhumita Basu
- Center for Cardiovascular Research, 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
| | - Vidu Garg
- Center for Cardiovascular Research, 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
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
11
|
Escalera A, Pascual I, Hernandez-Vaquero D, Formica F, Casares J, Diaz R, Alvarez R, Callejo F, Morales C, Moris C, Silva J. Association of the Surgical Technique With the Structural Valve Deterioration of a Bioprosthesis: A Prospective Cohort Study. Semin Thorac Cardiovasc Surg 2022; 35:647-655. [PMID: 35777692 DOI: 10.1053/j.semtcvs.2022.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 11/11/2022]
Abstract
The Trifecta aortic valve is a prosthesis with externally mounted leaflets and a stent which may be deformed during implant. Our aim was to know if the use of the holder as a protection device during the knotting has an impact on the incidence of structural valve deterioration (SVD) or endocarditis. Prospective cohort study where all patients who underwent aortic valve replacement with a Trifecta aortic valve between 2013 and 2018 were included. The use of the holder as a protection device was collected in a database. Propensity-score matched methods were used and analyses were based on competing events. Death without SVD or prosthesis replacement not due to SVD was considered competing events. 782 patients were included, 352 pairs after the matching. Rates of SVD at 5 and 8 years were 5.8% (95% CI 3.5-8.7) and 13.6% (95% CI 9.2-18.9) in the group without holder and 2.3% (95% CI 1-4.5) and 7% (95% CI 4.2-10.8) in the group with holder; sHR = 0.49 (95% CI 0.27-0.86; P = 0.015). The risk of endocarditis at 8 years was 4.8% (95% CI 2.8-7.4) in the group without holder and 2.3% (95% CI 1.1-4.3) in the group with holder, sHR = 0.49 (95% CI 0.21-1.15, P = 0.1). The use of holder as a protection device during the knotting of the Trifecta aortic valve is associated with less risk of SVD.
Collapse
Affiliation(s)
- Alain Escalera
- Cardiac Surgery Department, Central University Hospital of Asturias. Oviedo, Spain
| | - Isaac Pascual
- Department of Cardiology, Central University Hospital of Asturias. Oviedo, Spain; Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Daniel Hernandez-Vaquero
- Cardiac Surgery Department, Central University Hospital of Asturias. Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.
| | - Francesco Formica
- University of Parma, Department of Medicine and Surgery, Cardiac Surgery Clinic, Parma, Italy
| | - Julio Casares
- Department of Cardiology, Medical Center of Asturias, Oviedo, Spain
| | - Rocio Diaz
- Cardiac Surgery Department, Central University Hospital of Asturias. Oviedo, Spain
| | - Ruben Alvarez
- Cardiac Surgery Department, Central University Hospital of Asturias. Oviedo, Spain
| | - Francisco Callejo
- Cardiac Surgery Department, Central University Hospital of Asturias. Oviedo, Spain
| | - Carlos Morales
- Cardiac Surgery Department, Central University Hospital of Asturias. Oviedo, Spain
| | - Cesar Moris
- Department of Cardiology, Central University Hospital of Asturias. Oviedo, Spain; Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Jacobo Silva
- Cardiac Surgery Department, Central University Hospital of Asturias. Oviedo, Spain; Department of Medicine, University of Oviedo, Oviedo, Spain
| |
Collapse
|
12
|
Ground M, Waqanivavalagi S, Park YE, Callon K, Walker R, Milsom P, Cornish J. Fibroblast growth factor 2 inhibits myofibroblastic activation of valvular interstitial cells. PLoS One 2022; 17:e0270227. [PMID: 35714127 PMCID: PMC9205485 DOI: 10.1371/journal.pone.0270227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022] Open
Abstract
Heart valve disease is a growing problem worldwide. Though very common in older adults, the mechanisms behind the development of the disease aren't well understood, and at present the only therapeutic option is valve replacement. Valvular interstitial cells (VICs) may hold the answer. These cells can undergo pathological differentiation into contractile myofibroblasts or osteoblasts, leading to thickening and calcification of the valve tissue. Our study aimed to characterise the effect of fibroblast growth factor 2 (FGF-2) on the differentiation potential of VICs. We isolated VICs from diseased human valves and treated these cells with FGF-2 and TGF-β to elucidate effect of these growth factors on several myofibroblastic outcomes, in particular immunocytochemistry and gene expression. We used TGF-β as a positive control for myofibroblastic differentiation. We found that FGF-2 promotes a 'quiescent-type' morphology and inhibits the formation of α-smooth muscle actin positive myofibroblasts. FGF-2 reduced the calcification potential of VICs, with a marked reduction in the number of calcific nodules. FGF-2 interrupted the 'canonical' TGF-β signalling pathway, reducing the nuclear translocation of the SMAD2/3 complex. The panel of genes assayed revealed that FGF-2 promoted a quiescent-type pattern of gene expression, with significant downregulations in typical myofibroblast markers α smooth muscle actin, extracellular matrix proteins, and scleraxis. We did not see evidence of osteoblast differentiation: neither matrix-type calcification nor changes in osteoblast associated gene expression were observed. Our findings show that FGF-2 can reverse the myofibroblastic phenotype of VICs isolated from diseased valves and inhibit the calcification potential of these cells.
Collapse
Affiliation(s)
- Marcus Ground
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Steve Waqanivavalagi
- Green Lane Cardiothoracic Surgery Unit, Auckland City Hospital, Auckland District Health Board, Grafton, New Zealand
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, New Zealand
| | - Young-Eun Park
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, New Zealand
| | - Karen Callon
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, New Zealand
| | - Robert Walker
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Paget Milsom
- Green Lane Cardiothoracic Surgery Unit, Auckland City Hospital, Auckland District Health Board, Grafton, New Zealand
| | - Jillian Cornish
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, New Zealand
| |
Collapse
|
13
|
The Haemodynamic and Pathophysiological Mechanisms of Calcific Aortic Valve Disease. Biomedicines 2022; 10:biomedicines10061317. [PMID: 35740339 PMCID: PMC9220142 DOI: 10.3390/biomedicines10061317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
|
14
|
Vadana M, Cecoltan S, Ciortan L, Macarie RD, Mihaila AC, Tucureanu MM, Gan AM, Simionescu M, Manduteanu I, Droc I, Butoi E. Parathyroid Hormone Induces Human Valvular Endothelial Cells Dysfunction That Impacts the Osteogenic Phenotype of Valvular Interstitial Cells. Int J Mol Sci 2022; 23:ijms23073776. [PMID: 35409134 PMCID: PMC8998852 DOI: 10.3390/ijms23073776] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/25/2022] [Accepted: 03/27/2022] [Indexed: 11/16/2022] Open
Abstract
Parathyroid hormone (PTH) is a key regulator of calcium, phosphate and vitamin D metabolism. Although it has been reported that aortic valve calcification was positively associated with PTH, the pathophysiological mechanisms and the direct effects of PTH on human valvular cells remain unclear. Here we investigated if PTH induces human valvular endothelial cells (VEC) dysfunction that in turn could impact the switch of valvular interstitial cells (VIC) to an osteoblastic phenotype. Human VEC exposed to PTH were analyzed by qPCR, western blot, Seahorse, ELISA and immunofluorescence. Our results showed that exposure of VEC to PTH affects VEC metabolism and functions, modifications that were accompanied by the activation of p38MAPK and ERK1/2 signaling pathways and by an increased expression of osteogenic molecules (BMP-2, BSP, osteocalcin and Runx2). The impact of dysfunctional VEC on VIC was investigated by exposure of VIC to VEC secretome, and the results showed that VIC upregulate molecules associated with osteogenesis (BMP-2/4, osteocalcin and TGF-β1) and downregulate collagen I and III. In summary, our data show that PTH induces VEC dysfunction, which further stimulates VIC to differentiate into a pro-osteogenic pathological phenotype related to the calcification process. These findings shed light on the mechanisms by which PTH participates in valve calcification pathology and suggests that PTH and the treatment of hyperparathyroidism represent a therapeutic strategy to reduce valvular calcification.
Collapse
Affiliation(s)
- Mihaela Vadana
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Sergiu Cecoltan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Letitia Ciortan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Razvan D. Macarie
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Andreea C. Mihaila
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Monica M. Tucureanu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Ana-Maria Gan
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Maya Simionescu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Ileana Manduteanu
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
| | - Ionel Droc
- Cardiovascular Surgery Department, Central Military Hospital, 010825 Bucharest, Romania;
| | - Elena Butoi
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (M.V.); (S.C.); (L.C.); (R.D.M.); (A.C.M.); (M.M.T.); (A.-M.G.); (M.S.); (I.M.)
- Correspondence:
| |
Collapse
|
15
|
Ahuja N, Hiltabidle MS, Rajasekhar H, Voss S, Lu SZ, Barlow HR, Cowdin MA, Daniel E, Vaddaraju V, Anandakumar T, Black E, Cleaver O, Maynard C. Endothelial Cyp26b1 restrains murine heart valve growth during development. Dev Biol 2022; 486:81-95. [DOI: 10.1016/j.ydbio.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 11/28/2022]
|
16
|
Xiong T, Han S, Pu L, Zhang TC, Zhan X, Fu T, Dai YH, Li YX. Bioinformatics and Machine Learning Methods to Identify FN1 as a Novel Biomarker of Aortic Valve Calcification. Front Cardiovasc Med 2022; 9:832591. [PMID: 35295271 PMCID: PMC8918776 DOI: 10.3389/fcvm.2022.832591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
AimThe purpose of this study was to identify potential diagnostic markers for aortic valve calcification (AVC) and to investigate the function of immune cell infiltration in this disease.MethodsThe AVC data sets were obtained from the Gene Expression Omnibus. The identification of differentially expressed genes (DEGs) and the performance of functional correlation analysis were carried out using the R software. To explore hub genes related to AVC, a protein–protein interaction network was created. Diagnostic markers for AVC were then screened and verified using the least absolute shrinkage and selection operator, logistic regression, support vector machine-recursive feature elimination algorithms, and hub genes. The infiltration of immune cells into AVC tissues was evaluated using CIBERSORT, and the correlation between diagnostic markers and infiltrating immune cells was analyzed. Finally, the Connectivity Map database was used to forecast the candidate small molecule drugs that might be used as prospective medications to treat AVC.ResultsA total of 337 DEGs were screened. The DEGs that were discovered were mostly related with atherosclerosis and arteriosclerotic cardiovascular disease, according to the analyses. Gene sets involved in the chemokine signaling pathway and cytokine–cytokine receptor interaction were differently active in AVC compared with control. As the diagnostic marker for AVC, fibronectin 1 (FN1) (area the curve = 0.958) was discovered. Immune cell infiltration analysis revealed that the AVC process may be mediated by naïve B cells, memory B cells, plasma cells, activated natural killer cells, monocytes, and macrophages M0. Additionally, FN1 expression was associated with memory B cells, M0 macrophages, activated mast cells, resting mast cells, monocytes, and activated natural killer cells. AVC may be reversed with the use of yohimbic acid, the most promising small molecule discovered so far.ConclusionFN1 can be used as a diagnostic marker for AVC. It has been shown that immune cell infiltration is important in the onset and progression of AVC, which may benefit in the improvement of AVC diagnosis and treatment.
Collapse
Affiliation(s)
- Tao Xiong
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Shen Han
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Lei Pu
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Tian-Chen Zhang
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Xu Zhan
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Fu
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Ying-Hai Dai
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Ya-Xiong Li
- Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Yan'an Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
- *Correspondence: Ya-Xiong Li ;
| |
Collapse
|
17
|
Moore KS, Moore R, Fulmer DB, Guo L, Gensemer C, Stairley R, Glover J, Beck TC, Morningstar JE, Biggs R, Muhkerjee R, Awgulewitsch A, Norris RA. DCHS1, Lix1L, and the Septin Cytoskeleton: Molecular and Developmental Etiology of Mitral Valve Prolapse. J Cardiovasc Dev Dis 2022; 9:62. [PMID: 35200715 PMCID: PMC8874669 DOI: 10.3390/jcdd9020062] [Citation(s) in RCA: 6] [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: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Mitral valve prolapse (MVP) is a common cardiac valve disease that often progresses to serious secondary complications requiring surgery. MVP manifests as extracellular matrix disorganization and biomechanically incompetent tissues in the adult setting. However, MVP has recently been shown to have a developmental basis, as multiple causal genes expressed during embryonic development have been identified. Disease phenotypes have been observed in mouse models with human MVP mutations as early as birth. This study focuses on the developmental function of DCHS1, one of the first genes to be shown as causal in multiple families with non-syndromic MVP. By using various biochemical techniques as well as mouse and cell culture models, we demonstrate a unique link between DCHS1-based cell adhesions and the septin-actin cytoskeleton through interactions with cytoplasmic protein Lix1-Like (LIX1L). This DCHS1-LIX1L-SEPT9 axis interacts with and promotes filamentous actin organization to direct cell-ECM alignment and valve tissue shape.
Collapse
Affiliation(s)
- Kelsey S. Moore
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Reece Moore
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Diana B. Fulmer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Lilong Guo
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Cortney Gensemer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Rebecca Stairley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Janiece Glover
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Tyler C. Beck
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Jordan E. Morningstar
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Rachel Biggs
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Rupak Muhkerjee
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Alexander Awgulewitsch
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| |
Collapse
|
18
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
19
|
Tandon I, Ozkizilcik A, Ravishankar P, Balachandran K. Aortic valve cell microenvironment: Considerations for developing a valve-on-chip. BIOPHYSICS REVIEWS 2021; 2:041303. [PMID: 38504720 PMCID: PMC10903420 DOI: 10.1063/5.0063608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/15/2021] [Indexed: 03/21/2024]
Abstract
Cardiac valves are sophisticated, dynamic structures residing in a complex mechanical and hemodynamic environment. Cardiac valve disease is an active and progressive disease resulting in severe socioeconomic burden, especially in the elderly. Valve disease also leads to a 50% increase in the possibility of associated cardiovascular events. Yet, valve replacement remains the standard of treatment with early detection, mitigation, and alternate therapeutic strategies still lacking. Effective study models are required to further elucidate disease mechanisms and diagnostic and therapeutic strategies. Organ-on-chip models offer a unique and powerful environment that incorporates the ease and reproducibility of in vitro systems along with the complexity and physiological recapitulation of the in vivo system. The key to developing effective valve-on-chip models is maintaining the cell and tissue-level microenvironment relevant to the study application. This review outlines the various components and factors that comprise and/or affect the cell microenvironment that ought to be considered while constructing a valve-on-chip model. This review also dives into the advancements made toward constructing valve-on-chip models with a specific focus on the aortic valve, that is, in vitro studies incorporating three-dimensional co-culture models that incorporate relevant extracellular matrices and mechanical and hemodynamic cues.
Collapse
Affiliation(s)
- Ishita Tandon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Asya Ozkizilcik
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Prashanth Ravishankar
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas 72701, USA
| |
Collapse
|
20
|
Deb N, Lacerda CMR. Valvular Endothelial Cell Response to the Mechanical Environment-A Review. Cell Biochem Biophys 2021; 79:695-709. [PMID: 34661855 DOI: 10.1007/s12013-021-01039-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/02/2021] [Indexed: 01/08/2023]
Abstract
Heart valve leaflets are complex structures containing valve endothelial cells, interstitial cells, and extracellular matrix. Heart valve endothelial cells sense mechanical stimuli, and communicate amongst themselves and the surrounding cells and extracellular matrix to maintain tissue homeostasis. In the presence of abnormal mechanical stimuli, endothelial cell communication is triggered in defense and such processes may eventually lead to cardiac disease progression. This review focuses on the role of mechanical stimuli on heart valve endothelial surfaces-from heart valve development and maintenance of tissue integrity to disease progression with related signal pathways involved in this process.
Collapse
Affiliation(s)
- Nandini Deb
- Jasper Department of Chemical Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, 75799, TX, US
| | - Carla M R Lacerda
- Jasper Department of Chemical Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, 75799, TX, US.
| |
Collapse
|
21
|
Zebhi B, Lazkani M, Bark D. Calcific Aortic Stenosis-A Review on Acquired Mechanisms of the Disease and Treatments. Front Cardiovasc Med 2021; 8:734175. [PMID: 34604358 PMCID: PMC8486019 DOI: 10.3389/fcvm.2021.734175] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
Calcific aortic stenosis is a progressive disease that has become more prevalent in recent decades. Despite advances in research to uncover underlying biomechanisms, and development of new generations of prosthetic valves and replacement techniques, management of calcific aortic stenosis still comes with unresolved complications. In this review, we highlight underlying molecular mechanisms of acquired aortic stenosis calcification in relation to hemodynamics, complications related to the disease, diagnostic methods, and evolving treatment practices for calcific aortic stenosis.
Collapse
Affiliation(s)
- Banafsheh Zebhi
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Mohamad Lazkani
- Medical Center of the Rockies, University of Colorado Health, Loveland, CO, United States
| | - David Bark
- Department of Pediatrics, Washington University in Saint Louis, Saint Louis, MO, United States.,Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, United States
| |
Collapse
|
22
|
Deb N, Ali MS, Mathews A, Chang YW, Lacerda CM. Shear type and magnitude affect aortic valve endothelial cell morphology, orientation, and differentiation. Exp Biol Med (Maywood) 2021; 246:2278-2289. [PMID: 34260291 DOI: 10.1177/15353702211023359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Valvular endothelial cells line the outer layer of heart valves and can withstand shear forces caused by blood flow. In contrast to vascular endothelial cells, there is limited amount of research over valvular endothelial cells. For this reason, the exact physiologic behavior of valvular endothelial cells is unclear. Prior studies have concluded that valvular endothelial cells align perpendicularly to the direction of blood flow, while vascular endothelial cells align parallel to blood flow. Other studies have suggested that different ranges of shear stress uniquely impact the behavior of valvular endothelial cells. The goal of this study was to characterize the response of valvular endothelial cell under different types, magnitudes, and durations of shear stress. In this work, the results demonstrated that with increased shear rate and duration of exposure, valvular endothelial cells no longer possessed the traditional cuboidal morphology. Instead through the change in cell circularity and aspect ratio, valvular endothelial cells aligned in an organized manner. In addition, different forms of shear exposure caused the area and circularity of valvular endothelial cells to decrease while inducing mesenchymal transformation validated through αSMA and TGFβ1 expression. This is the first investigation showing that valvular endothelial cells alignment is not as straightforward as once thought (perpendicular to flow). Different types and magnitudes of shear induce different local behaviors. This is also the first demonstration of valvular endothelial cells undergoing EndMT without chemical inducers on a soft surface in vitro. Findings from this study provide insights to understanding the pathophysiology of valvular endothelial cells which can potentially propel future artificial engineered heart valves.
Collapse
Affiliation(s)
- Nandini Deb
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Mir S Ali
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ashley Mathews
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Ya-Wen Chang
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Carla Mr Lacerda
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| |
Collapse
|
23
|
Soltany Sadrabadi M, Hedayat M, Borazjani I, Arzani A. Fluid-structure coupled biotransport processes in aortic valve disease. J Biomech 2021; 117:110239. [PMID: 33515904 DOI: 10.1016/j.jbiomech.2021.110239] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/22/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022]
Abstract
Biological transport processes near the aortic valve play a crucial role in calcific aortic valve disease initiation and bioprosthetic aortic valve thrombosis. Hemodynamics coupled with the dynamics of the leaflets regulate these transport patterns. Herein, two-way coupled fluid-structure interaction (FSI) simulations of a 2D bicuspid aortic valve and a 3D mechanical heart valve were performed and coupled with various convective mass transport models that represent some of the transport processes in calcification and thrombosis. Namely, five different continuum transport models were developed to study biochemicals that originate from the blood and the leaflets, as well as residence-time and flow stagnation. Low-density lipoprotein (LDL) and platelet activation were studied for their role in calcification and thrombosis, respectively. Coherent structures were identified using vorticity and Lagrangian coherent structures (LCS) for the 2D and 3D models, respectively. A very close connection between vortex structures and biochemical concentration patterns was shown where different vortices controlled the concentration patterns depending on the transport mechanism. Additionally, the relationship between leaflet concentration and wall shear stress was revealed. Our work shows that blood flow physics and coherent structures regulate the flow-mediated biological processes that are involved in aortic valve calcification and thrombosis, and therefore could be used in the design process to optimize heart valve replacement durability.
Collapse
Affiliation(s)
| | - Mohammadali Hedayat
- J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, TX, USA
| | - Iman Borazjani
- J. Mike Walker '66 Department of Mechanical Engineering, Texas A&M University, College Station, TX, USA
| | - Amirhossein Arzani
- Department of Mechanical Engineering, Northern Arizona University, Flagstaff, AZ, USA.
| |
Collapse
|
24
|
Hamza O, Kiss A, Kramer AM, Trojanek S, Abraham D, Acar E, Nagel F, Tretter VE, Kitzwögerer M, Podesser BK. Tenascin C promotes valvular remodeling in two large animal models of ischemic mitral regurgitation. Basic Res Cardiol 2020; 115:76. [PMID: 33258993 PMCID: PMC7716900 DOI: 10.1007/s00395-020-00837-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/25/2020] [Indexed: 01/03/2023]
Abstract
Ischemic mitral regurgitation (MR) is a frequent complication of myocardial infarction (MI) characterized by adverse remodeling both at the myocardial and valvular levels. Persistent activation of valvular endothelial cells leads to leaflet fibrosis through endothelial-to-mesenchymal transition (EMT). Tenascin C (TNC), an extracellular matrix glycoprotein involved in cardiovascular remodeling and fibrosis, was also identified in inducing epithelial-to-mesenchymal transition. In this study, we hypothesized that TNC also plays a role in the valvular remodeling observed in ischemic MR by contributing to valvular excess EMT. Moderate ischemic MR was induced by creating a posterior papillary muscle infarct (7 pigs and 7 sheep). Additional animals (7 pigs and 4 sheep) served as controls. Pigs and sheep were sacrificed after 6 weeks and 6 months, respectively. TNC expression was upregulated in the pig and sheep experiments at 6 weeks and 6 months, respectively, and correlated well with leaflet thickness (R = 0.68; p < 0.001 at 6 weeks, R = 0.84; p < 0.001 at 6 months). To confirm the translational potential of our findings, we obtained mitral valves from patients with ischemic cardiomyopathy presenting MR (n = 5). Indeed, TNC was also expressed in the mitral leaflets of these. Furthermore, TNC induced EMT in isolated porcine mitral valve endothelial cells (MVEC). Interestingly, Toll-like receptor 4 (TLR4) inhibition prevented TNC-mediated EMT in MVEC. We identified here for the first time a new contributor to valvular remodeling in ischemic MR, namely TNC, which induced EMT through TLR4. Our findings might set the path for novel therapeutic targets for preventing or limiting ischemic MR.
Collapse
Affiliation(s)
- Ouafa Hamza
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Anne-Margarethe Kramer
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Sandra Trojanek
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Abraham
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Eylem Acar
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Felix Nagel
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Cardiac Surgery, Karl Landsteiner University, St. Pölten, Austria
| | - Verena Eva Tretter
- Department of Anesthesia, General Intensive Care and Pain Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Department of Cardiac Surgery, Karl Landsteiner University, St. Pölten, Austria.
| |
Collapse
|
25
|
Side-dependent effect in the response of valve endothelial cells to bidirectional shear stress. Int J Cardiol 2020; 323:220-228. [PMID: 32858136 DOI: 10.1016/j.ijcard.2020.08.074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/10/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022]
Abstract
Endothelial cells covering the aortic and ventricular sides of the aortic valve leaflets are exposed to different stresses, in particular wall shear stress (WSS). Biomechanical stimuli actively regulate valve tissue structure and induce remodeling events leading to valve dysfunction. Endothelial to mesenchymal transformation (EndMT), for example, has been associated with aortic valve disease. The biomechanical response of cells at different sides of the leaflets has not been clearly characterized. To analyze the mechanical response of valve endothelial cells (VECs) we developed a unique fluid activation device that applies physiologically relevant pulsatile WSS. We characterized the morphology and function of adult porcine aortic VECs derived from the opposite sides of aortic valve leaflets following exposure to different pulsatile WSS. We found that elongation and orientation of cells in response to pulsatile WSS depends on their side of origin. Quantification of gene expression confirms phenotypic differences between aortic and ventricular VECs. Aortic VECs exposed to pulsatile WSS similar to that in vivo at the tip of aortic side of the valve leaflet upregulated pro-EndMT (ACTA2, Snail, TGFβ1) and inflammation (ICAM-1, VCAM-1) genes, whereas expression of endothelial markers like PECAM-1 was decreased. Conversely, ventricular-VECs showed strong increase of PECAM-1 expression and no activation of pro-EndMT marker. Finally, we found that stress-induced genes are upregulated in both cell types, at higher levels in ventricular compared to aortic VECs. Application of physiological shear stress levels using a fluid activation device therefore reveals functional differences in VECs derived from opposite sides of the aortic valve leaflets.
Collapse
|
26
|
Kandasamy M, Anusuyadevi M, Aigner KM, Unger MS, Kniewallner KM, de Sousa DMB, Altendorfer B, Mrowetz H, Bogdahn U, Aigner L. TGF-β Signaling: A Therapeutic Target to Reinstate Regenerative Plasticity in Vascular Dementia? Aging Dis 2020; 11:828-850. [PMID: 32765949 PMCID: PMC7390515 DOI: 10.14336/ad.2020.0222] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/22/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular dementia (VaD) is the second leading form of memory loss after Alzheimer's disease (AD). Currently, there is no cure available. The etiology, pathophysiology and clinical manifestations of VaD are extremely heterogeneous, but the impaired cerebral blood flow (CBF) represents a common denominator of VaD. The latter might be the result of atherosclerosis, amyloid angiopathy, microbleeding and micro-strokes, together causing blood-brain barrier (BBB) dysfunction and vessel leakage, collectively originating from the consequence of hypertension, one of the main risk factors for VaD. At the histopathological level, VaD displays abnormal vascular remodeling, endothelial cell death, string vessel formation, pericyte responses, fibrosis, astrogliosis, sclerosis, microglia activation, neuroinflammation, demyelination, white matter lesions, deprivation of synapses and neuronal loss. The transforming growth factor (TGF) β has been identified as one of the key molecular factors involved in the aforementioned various pathological aspects. Thus, targeting TGF-β signaling in the brain might be a promising therapeutic strategy to mitigate vascular pathology and improve cognitive functions in patients with VaD. This review revisits the recent understanding of the role of TGF-β in VaD and associated pathological hallmarks. It further explores the potential to modulate certain aspects of VaD pathology by targeting TGF-β signaling.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India.
| | - Muthuswamy Anusuyadevi
- Molecular Gerontology Group, Department of Biochemistry, School of Life Sciences, Bharathidhasan University, Tiruchirappalli, Tamil Nadu, India.
| | - Kiera M Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Kathrin M Kniewallner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Diana M Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Ulrich Bogdahn
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Velvio GmbH, Regensburg, Germany.
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
27
|
de Oliveira DMC, Abdullah N, Green NC, Espino DM. Biomechanical Assessment of Bicuspid Aortic Valve Phenotypes: A Fluid-Structure Interaction Modelling Approach. Cardiovasc Eng Technol 2020; 11:431-447. [PMID: 32519086 DOI: 10.1007/s13239-020-00469-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Bicuspid aortic valve (BAV) is a congenital heart malformation with phenotypic heterogeneity. There is no prior computational study that assesses the haemodynamic and valve mechanics associated with BAV type 2 against a healthy tricuspid aortic valve (TAV) and other BAV categories. METHODS A proof-of-concept study incorporating three-dimensional fluid-structure interaction (FSI) models with idealised geometries (one TAV and six BAVs, namely type 0 with lateral and anterior-posterior orientations, type 1 with R-L, N-R and N-L leaflet fusion and type 2) has been developed. Transient physiological boundary conditions have been applied and simulations were run using an Arbitrary Lagrangian-Eulerian formulation. RESULTS Our results showed the presence of abnormal haemodynamics in the aorta and abnormal valve mechanics: type 0 BAVs yielded the best haemodynamical and mechanical outcomes, but cusp stress distribution varied with valve orifice orientation, which can be linked to different cusp calcification location onset; type 1 BAVs gave rise to similar haemodynamics and valve mechanics, regardless of raphe position, but this position altered the location of abnormal haemodynamic features; finally, type 2 BAV constricted the majority of blood flow, exhibiting the most damaging haemodynamic and mechanical repercussions when compared to other BAV phenotypes. CONCLUSION The findings of this proof-of-concept work suggest that there are specific differences across haemodynamics and valve mechanics associated with BAV phenotypes, which may be critical to subsequent processes associated with their pathophysiology processes.
Collapse
Affiliation(s)
- Diana M C de Oliveira
- Department of Mechanical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Nazirul Abdullah
- Department of Mechanical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Naomi C Green
- Department of Mechanical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Daniel M Espino
- Department of Mechanical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
28
|
Bidar E, Folliguet T, Kluin J, Muneretto C, Parolari A, Barili F, Suwalski P, Bonaros N, Punjabi P, Sadaba R, De Bonis M, Al-Attar N, Obadia JF, Czerny M, Shrestha M, Zegdi R, Natour E, Lorusso R. Postimplant biological aortic prosthesis degeneration: challenges in transcatheter valve implants. Eur J Cardiothorac Surg 2019; 55:191-200. [PMID: 30541101 DOI: 10.1093/ejcts/ezy391] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/15/2018] [Indexed: 12/29/2022] Open
Abstract
Surgical aortic valve replacement (SAVR) is highly effective and can be achieved with relatively low risk in patients with severe aortic stenosis. Bioprostheses have been used most frequently during the past 60 years. However, the function of biological valves usually declines after 10-15 years from implant when structural valve degeneration occurs often mandating a reoperation once valve dysfunction becomes haemodynamically significant. Known for many years by surgeons and cardiologists taking care of patients with SAVR, the issue of postimplant structural valve degeneration has been recently highlighted also in patients with transcatheter aortic valve implant (TAVI). There is growing concern that TAVI valves exhibit structural valve degeneration due to inherent challenges of the deployment mode. The impact on postimplant degeneration of TAVI valves compared to SAVR has still to be understood and defined. Based on the ongoing process of expanding TAVI indications, several potential shortcomings and caveats, learned during the last 60 years of SAVR experience, should be taken into consideration to refine this technique.
Collapse
Affiliation(s)
- Elham Bidar
- Department of Cardio-Thoracic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Thierry Folliguet
- Centre Hospitalo-Universitaire Brabois ILCV, Hôpital Henri Mondor, Division of Cardio Thoracic Surgery and Transplantation, Université Paris 12 UPEC, France
| | - Jolanda Kluin
- Department of Cardio-Thoracic Surgery, Academic Medical Center, Amsterdam, Netherlands
| | - Claudio Muneretto
- Cardiac Surgery Unit, University of Brescia Medical School, Brescia, Italy
| | - Alessandro Parolari
- Cardiac Surgery and Translational Research Units, IRCCS, Policlinico S. Donato, University of Milan, Milan, Italy
| | - Fabio Barili
- Department of Cardiac Surgery, S. Croce Hospital, Cuneo, Italy
| | - Piotr Suwalski
- Department of Cardiac Surgery, Central Clinical Hospital of the Ministry of Interior and Administration, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Nikolaos Bonaros
- Department of Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Prakash Punjabi
- Department of Cardio-Thoracic Surgery, Imperial College Healthcare NHS Trust, Imperial College School of Medicine, London, UK
| | - Rafa Sadaba
- Department of Cardiac Surgery, Hospital de Navarra, Pamplona, Spain
| | - Michele De Bonis
- Department of Cardiac Surgery, S. Raffaele University Hospital, Milan, Italy
| | - Nawwar Al-Attar
- Department of Cardiac Surgery, Golden Jubilee National Hospital, Glasgow, UK
| | - Jean Francois Obadia
- Department of Cardio-Thoracic Surgery, Hôpital Cardiothoracique Louis Pradel, Lyon, France
| | - Martin Czerny
- Department of Cardio-Vascular Surgery, University Hospital Freiburg, Freiburg, Germany
| | - Malakh Shrestha
- Department of Cardio-Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Rachid Zegdi
- Hôpital Européen Georges Pompidou, Paris, France
| | - Ehsan Natour
- Department of Cardio-Thoracic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Roberto Lorusso
- Department of Cardio-Thoracic Surgery, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
29
|
Odelin G, Faure E, Maurel-Zaffran C, Zaffran S. Krox20 Regulates Endothelial Nitric Oxide Signaling in Aortic Valve Development and Disease. J Cardiovasc Dev Dis 2019; 6:jcdd6040039. [PMID: 31684048 PMCID: PMC6955692 DOI: 10.3390/jcdd6040039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/25/2019] [Accepted: 10/31/2019] [Indexed: 12/18/2022] Open
Abstract
Among the aortic valve diseases, the bicuspid aortic valve (BAV) occurs when the aortic valve has two leaflets (cusps), rather than three, and represents the most common form of congenital cardiac malformation, affecting 1–2% of the population. Despite recent advances, the etiology of BAV is poorly understood. We have recently shown that Krox20 is expressed in endothelial and cardiac neural crest derivatives that normally contribute to aortic valve development and that lack of Krox20 in these cells leads to aortic valve defects including partially penetrant BAV formation. Dysregulated expression of endothelial nitric oxide synthase (Nos3) is associated with BAV. To investigate the relationship between Krox20 and Nos3 during aortic valve development, we performed inter-genetic cross. While single heterozygous mice had normal valve formation, the compound Krox20+/−;Nos3+/− mice had BAV malformations displaying an in vivo genetic interaction between these genes for normal valve morphogenesis. Moreover, in vivo and in vitro experiments demonstrate that Krox20 directly binds to Nos3 proximal promoter to activate its expression. Our data suggests that Krox20 is a regulator of nitric oxide in endothelial-derived cells in the development of the aortic valve and concludes on the interaction of Krox20 and Nos3 in BAV formation.
Collapse
Affiliation(s)
- Gaëlle Odelin
- Aix Marseille University, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France.
| | - Emilie Faure
- Aix Marseille University, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France.
| | | | - Stéphane Zaffran
- Aix Marseille University, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France.
| |
Collapse
|
30
|
Development of calcific aortic valve disease: Do we know enough for new clinical trials? J Mol Cell Cardiol 2019; 132:189-209. [PMID: 31136747 DOI: 10.1016/j.yjmcc.2019.05.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/11/2019] [Accepted: 05/19/2019] [Indexed: 12/19/2022]
Abstract
Calcific aortic valve disease (CAVD), previously thought to represent a passive degeneration of the valvular extracellular matrix (VECM), is now regarded as an intricate multistage disorder with sequential yet intertangled and interacting underlying processes. Endothelial dysfunction and injury, initiated by disturbed blood flow and metabolic disorders, lead to the deposition of low-density lipoprotein cholesterol in the VECM further provoking macrophage infiltration, oxidative stress, and release of pro-inflammatory cytokines. Such changes in the valvular homeostasis induce differentiation of normally quiescent valvular interstitial cells (VICs) into synthetically active myofibroblasts producing excessive quantities of the VECM and proteins responsible for its remodeling. As a result of constantly ongoing degradation and re-deposition, VECM becomes disorganised and rigid, additionally potentiating myofibroblastic differentiation of VICs and worsening adaptation of the valve to the blood flow. Moreover, disrupted and excessively vascularised VECM is susceptible to the dystrophic calcification caused by calcium and phosphate precipitating on damaged collagen fibers and concurrently accompanied by osteogenic differentiation of VICs. Being combined, passive calcification and biomineralisation synergistically induce ossification of the aortic valve ultimately resulting in its mechanical incompetence requiring surgical replacement. Unfortunately, multiple attempts have failed to find an efficient conservative treatment of CAVD; however, therapeutic regimens and clinical settings have also been far from the optimal. In this review, we focused on interactions and transitions between aforementioned mechanisms demarcating ascending stages of CAVD, suggesting a predisposing condition (bicuspid aortic valve) and drug combination (lipid-lowering drugs combined with angiotensin II antagonists and cytokine inhibitors) for the further testing in both preclinical and clinical trials.
Collapse
|
31
|
Ali MS, Wang X, Lacerda CMR. The effect of physiological stretch and the valvular endothelium on mitral valve proteomes. Exp Biol Med (Maywood) 2019; 244:241-251. [PMID: 30722697 PMCID: PMC6425102 DOI: 10.1177/1535370219829006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/09/2019] [Indexed: 11/15/2022] Open
Abstract
IMPACT STATEMENT This work is important to the field of heart valve pathophysiology as it provides new insights into molecular markers of mechanically induced valvular degeneration as well as the protective role of the valvular endothelium. These discoveries reported here advance our current knowledge of the valvular endothelium and how its removal essentially takes valve leaflets into an environmental shock. In addition, it shows that static conditions represent a mild pathological state for valve leaflets, while 10% cyclic stretch provides valvular cell quiescence. These findings impact the field by informing disease stages and by providing potential new drug targets to reverse or slow down valvular change before it affects cardiac function.
Collapse
Affiliation(s)
- Mir S Ali
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Xinmei Wang
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Carla MR Lacerda
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| |
Collapse
|
32
|
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
|
33
|
Haataja TJK, Bernardi RC, Lecointe S, Capoulade R, Merot J, Pentikäinen U. Non-syndromic Mitral Valve Dysplasia Mutation Changes the Force Resilience and Interaction of Human Filamin A. Structure 2018; 27:102-112.e4. [PMID: 30344108 DOI: 10.1016/j.str.2018.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/24/2018] [Accepted: 09/18/2018] [Indexed: 01/03/2023]
Abstract
Filamin A (FLNa), expressed in endocardial endothelia during fetal valve morphogenesis, is key in cardiac development. Missense mutations in FLNa cause non-syndromic mitral valve dysplasia (FLNA-MVD). Here, we aimed to reveal the currently unknown underlying molecular mechanism behind FLNA-MVD caused by the FLNa P637Q mutation. The solved crystal structure of the FLNa3-5 P637Q revealed that this mutation causes only minor structural changes close to mutation site. These changes were observed to significantly affect FLNa's ability to transmit cellular force and to interact with its binding partner. The performed steered molecular dynamics simulations showed that significantly lower forces are needed to split domains 4 and 5 in FLNA-MVD than with wild-type FLNa. The P637Q mutation was also observed to interfere with FLNa's interactions with the protein tyrosine phosphatase PTPN12. Our results provide a crucial step toward understanding the molecular bases behind FLNA-MVD, which is critical for the development of drug-based therapeutics.
Collapse
Affiliation(s)
- Tatu J K Haataja
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland
| | - Rafael C Bernardi
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Simon Lecointe
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | | | - Jean Merot
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Ulla Pentikäinen
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, 40014 Jyväskylä, Finland; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Institute of Biomedicine, University of Turku, 20520 Turku, Finland; Turku Centre for Biotechnology, University of Turku, 20520 Turku, Finland.
| |
Collapse
|
34
|
Lee SH, Choi JH. Involvement of inflammatory responses in the early development of calcific aortic valve disease: lessons from statin therapy. Anim Cells Syst (Seoul) 2018; 22:390-399. [PMID: 30533261 PMCID: PMC6282465 DOI: 10.1080/19768354.2018.1528175] [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: 04/29/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common degenerative heart valve disease. Among the many risk factors for this disease are age, hypercholesterolemia, hypertension, smoking, type-2 diabetes, rheumatic fever, and chronic kidney disease. Since many of these overlap with risk factors for atherosclerosis, the molecular and cellular mechanisms of CAVD development have been presumed to be similar to those for atherogenesis. Thus, attempts have been made to evaluate the therapeutic efficacy of statins, representative anti-atherosclerosis drugs with lipid-lowering and anti-inflammatory effects, against CAVD. Unfortunately, statins have shown little or no effect on CAVD development. But some reports suggest that statins may prevent or reduce the development of early stage CAVD in which having calcification is absent or minimal. These results suggest that therapeutic approaches should differ according to the stage of disease, and that a precise understanding of the mechanism of aortic valve calcification is required to identify novel therapeutic targets for advanced CAVD. Given the involvement of inflammatory processes in the development and progression of CAVD, current therapeutic approaches for chronic inflammatory cardiovascular disease like atherosclerosis may help to prevent or minimize the early development of CAVD. In this review, we focus on several inflammatory cellular and molecular components involved in CAVD that might be considered drug targets for preventing CAVD.
Collapse
Affiliation(s)
- Seung Hyun Lee
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
35
|
Lee J, Estlack Z, Somaweera H, Wang X, Lacerda CMR, Kim J. A microfluidic cardiac flow profile generator for studying the effect of shear stress on valvular endothelial cells. LAB ON A CHIP 2018; 18:2946-2954. [PMID: 30123895 DOI: 10.1039/c8lc00545a] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
To precisely investigate the mechanobiological responses of valvular endothelial cells, we developed a microfluidic flow profile generator using a pneumatically-actuated micropump consisting of microvalves of various sizes. By controlling the closing pressures and the actuation times of these microvalves, we modulated the magnitude and frequency of the shear stress to mimic mitral and aortic inflow profiles with frequencies in the range of 0.8-2 Hz and shear stresses up to 20 dyn cm-2. To demonstrate this flow profile generator, aortic inflow with an average of 5.9 dyn cm-2 shear stress at a frequency of 1.2 Hz with a Reynolds number of 2.75, a Womersley number of 0.27, and an oscillatory shear index (OSI) value of 0.2 was applied to porcine aortic valvular endothelial cells (PAVECs) for mechanobiological studies. The cell alignment, cell elongation, and alpha-smooth muscle actin (αSMA) expression of PAVECs under perfusion, steady flow, and aortic inflow conditions were analyzed to determine their shear-induced cell migration and trans-differentiation. In this morphological and immunocytochemical study, we found that the PAVECs elongated and aligned themselves perpendicular to the directions of the steady flow and the aortic inflow. In contrast, under perfusion with a fluidic shear stress of 0.47 dyn cm-2, the PAVECs elongated and aligned themselves parallel to the direction of flow. The PAVECs exposed to the aortic inflow upregulated their αSMA-protein expression to a greater degree than those exposed to perfusion and steady flow. By comparing these results to those of previous studies of pulsatile flow, we also found that the ratio of positive to negative shear stress plays an important role in determining PAVECs' trans-differentiation and adaptation to flow. This microfluidic cardiac flow profile generator will enable future valvular mechanobiological studies to determine the roles of magnitude and frequency of shear stresses.
Collapse
Affiliation(s)
- Joohyung Lee
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- Alberto Giannoni
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
- Scuola Superiore Sant’Anna, Pisa, Italy
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
- Department of Twin Research and Genetic Epidemiology, King’s College London, UK
- National Centre for Cardiovascular Preventions and Outcomes, Institute of Cardiovascular Science, University College London, UK
| |
Collapse
|
37
|
Jung JJ, Jadbabaie F, Sadeghi MM. Molecular imaging of calcific aortic valve disease. J Nucl Cardiol 2018; 25:1148-1155. [PMID: 29359271 PMCID: PMC6054901 DOI: 10.1007/s12350-017-1158-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 11/27/2017] [Indexed: 12/13/2022]
Abstract
Calcific aortic valve disease (CAVD) can progress to symptomatic aortic stenosis in a subset of patients. The severity of aortic stenosis and the extent of valvular calcification can be evaluated readily by echocardiography, CT, and MRI using well-established imaging protocols. However, these techniques fail to address optimally other important aspects of CAVD, including the propensity for disease progression, risk of complications in asymptomatic patients, and the effect of therapeutic interventions on valvular biology. These gaps may be addressed by molecular imaging targeted at key biological processes such as inflammation, remodeling, and calcification that mediate the development and progression of CAVD. In this review, recent advances in valvular molecular imaging, including 18F-fluorodeoxyglucose (FDG) and 18F-sodium fluoride (NaF) PET, and matrix metalloproteinase-targeted SPECT imaging in the preclinical and clinical settings are presented and discussed.
Collapse
Affiliation(s)
- Jae-Joon Jung
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, USA
- Yale Cardiovascular Research Center, 300 George Street, #770G, New Haven, CT, 06511, USA
| | - Farid Jadbabaie
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Mehran M Sadeghi
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, USA.
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA.
- Yale Cardiovascular Research Center, 300 George Street, #770G, New Haven, CT, 06511, USA.
| |
Collapse
|
38
|
Geng X, Cha B, Mahamud MR, Srinivasan RS. Intraluminal valves: development, function and disease. Dis Model Mech 2018; 10:1273-1287. [PMID: 29125824 PMCID: PMC5719258 DOI: 10.1242/dmm.030825] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The circulatory system consists of the heart, blood vessels and lymphatic vessels, which function in parallel to provide nutrients and remove waste from the body. Vascular function depends on valves, which regulate unidirectional fluid flow against gravitational and pressure gradients. Severe valve disorders can cause mortality and some are associated with severe morbidity. Although cardiac valve defects can be treated by valve replacement surgery, no treatment is currently available for valve disorders of the veins and lymphatics. Thus, a better understanding of valves, their development and the progression of valve disease is warranted. In the past decade, molecules that are important for vascular function in humans have been identified, with mouse studies also providing new insights into valve formation and function. Intriguing similarities have recently emerged between the different types of valves concerning their molecular identity, architecture and development. Shear stress generated by fluid flow has also been shown to regulate endothelial cell identity in valves. Here, we review our current understanding of valve development with an emphasis on its mechanobiology and significance to human health, and highlight unanswered questions and translational opportunities.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Md Riaj Mahamud
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA .,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
39
|
Castellanos G, Nasim S, Almora DM, Rath S, Ramaswamy S. Stem Cell Cytoskeletal Responses to Pulsatile Flow in Heart Valve Tissue Engineering Studies. Front Cardiovasc Med 2018; 5:58. [PMID: 29922678 PMCID: PMC5996090 DOI: 10.3389/fcvm.2018.00058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/15/2018] [Indexed: 01/12/2023] Open
Abstract
Heart valve replacement options remain exceedingly limited for pediatric patients because they cannot accommodate somatic growth. To overcome this shortcoming, heart valve tissue engineering using human bone marrow stem cells (HBMSCs) has been considered a potential solution to the treatment of critical congenital valvular defects. The mechanical environments during in vitro culture are key regulators of progenitor cell fate. Here, we report on alterations in HBMSCs, specifically in their actin cytoskeleton and their nucleus under fluid-induced shear stresses of relevance to heart valves. HBMSCs were seeded in microfluidic channels and were exposed to the following conditions: pulsatile shear stress (PSS), steady shear stress (SS), and no flow controls (n = 4/group). Changes to the actin filament structure were monitored and subsequent gene expression was evaluated. A significant increase (p < 0.05) in the number of actin filaments, filament density and angle (between 30° and 84°), and conversely a significant decrease (p < 0.05) in the length of the filaments were observed when the HBMSCs were exposed to PSS for 48 h compared to SS and no flow conditions. No significant differences in nuclear shape were observed among the groups (p > 0.05). Of particular relevance to valvulogenesis, klf2a, a critical gene in valve development, was significantly expressed only by the PSS group (p < 0.05). We conclude that HBMSCs respond to PSS by alterations to their actin filament structure that are distinct from SS and no flow conditions. These changes coupled with the subsequent gene expression findings suggest that at the cellular level, the immediate effect of PSS is to initiate a unique set of quantifiable cytoskeletal events (increased actin filament number, density and angle, but decrease in filament length) in stem cells, which could be useful in the fine-tuning of in vitro protocols in heart valve tissue engineering.
Collapse
Affiliation(s)
- Glenda Castellanos
- Tissue Engineered Mechanics Imaging and Materials Laboratory, Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Sana Nasim
- Tissue Engineered Mechanics Imaging and Materials Laboratory, Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Denise M Almora
- Tissue Engineered Mechanics Imaging and Materials Laboratory, Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Sasmita Rath
- Tissue Engineered Mechanics Imaging and Materials Laboratory, Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Sharan Ramaswamy
- Tissue Engineered Mechanics Imaging and Materials Laboratory, Biomedical Engineering, Florida International University, Miami, FL, United States
| |
Collapse
|
40
|
Richards JM, Kunitake JA, Hunt HB, Wnorowski AN, Lin DW, Boskey AL, Donnelly E, Estroff LA, Butcher JT. Crystallinity of hydroxyapatite drives myofibroblastic activation and calcification in aortic valves. Acta Biomater 2018; 71:24-36. [PMID: 29505892 DOI: 10.1016/j.actbio.2018.02.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/15/2018] [Accepted: 02/22/2018] [Indexed: 12/30/2022]
Abstract
Calcific aortic valve disease (CAVD) is an inexorably degenerative pathology characterized by progressive calcific lesion formation on the valve leaflets. The interaction of valvular cells in advanced lesion environments is not well understood yet highly relevant as clinically detectable CAVD exhibits calcifications composed of non-stoichiometric hydroxyapatite (HA). In this study, Fourier transform infrared spectroscopic imaging was used to spatially analyze mineral properties as a function of disease progression. Crystallinity (size and perfection) increased with increased valve calcification. To study the relationship between crystallinity and cellular behavior in CAVD, valve cells were seeded into 3D mineral-rich collagen gels containing synthetic HA particles, which had varying crystallinities. Lower crystallinity HA drove myofibroblastic activation in both valve interstitial and endothelial cells, as well as osteoblastic differentiation in interstitial cells. Additionally, calcium accumulation within gels depended on crystallinity, and apoptosis was insufficient to explain differences in HA-driven cellular activity. The protective nature of endothelial cells against interstitial cell activation and calcium accumulation was completely inhibited in the presence of less crystalline HA particles. Elucidating valve cellular behavior post-calcification is of vital importance to better predict and treat clinical pathogenesis, and mineral-containing hydrogel models provide a unique 3D platform to evaluate valve cell responses to a later stage of valve disease. STATEMENT OF SIGNIFICANCE We implement a 3D in vitro platform with embedded hydroxyapatite (HA) nanoparticles to investigate the interaction between valve interstitial cells, valve endothelial cells, and a mineral-rich extracellular environment. HA nanoparticles were synthesized based on analysis of the mineral properties of calcific regions of diseased human aortic valves. Our findings indicate that crystallinity of HA drives activation and differentiation in interstitial and endothelial cells. We also show that a mineralized environment blocks endothelial protection against interstitial cell calcification. Our HA-containing hydrogel model provides a unique 3D platform to evaluate valve cell responses to a mineralized ECM. This study additionally lays the groundwork to capture the diversity of mineral properties in calcified valves, and link these properties to progression of the disease.
Collapse
|
41
|
Zhang Q, Gao B, Yu C. The Effects of Left Ventricular Assist Device Support Level on the Biomechanical States of Aortic Valve. Med Sci Monit 2018; 24:2003-2017. [PMID: 29618718 PMCID: PMC5900803 DOI: 10.12659/msm.906903] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background Although aortic valve disease caused by left ventricular assist device (LVAD) support has attracted more and more attention, the precise biomechanical effects of LVAD support level on the aortic valve are still unclear. Material/Methods A structural finite element models study was conducted using an ideal aortic valve geometric model. Four different study conditions were designed, according to the reduction of the open duration of the aortic valve. The isotropic hyperelastic constitutive equation was chosen to reflect the mechanical property of the leaflets. The distribution of the stress, strain, and transient dynamics of the leaflet were calculated. Results Along with the increase of LVAD support level, the open duration of the aortic valve was also reduced by the increase of LVAD support (low support level case 0.23 seconds versus middle support level case 0.2 seconds versus high support level case 0.14 seconds). Moreover, along with the increase of support mode of LVAD, the von Mises stress in most leaflet areas was increased from the low stress level (0–0.4 MPa) to the middle region (0.4–0.8 MPa). Once the leaflets were continuously closed, the high stress level (larger than 0.8 MPa) was observed. In contrast, the support level of LVAD only had slight effects on the distribution of von Mises strain. According to the aforementioned results, maintaining the open duration of aortic valve longer than 0.2 seconds could achieve better performance of biomechanical states of leaflets. Conclusions This study could provide useful information on the determination of optimal LVAD support strategy.
Collapse
Affiliation(s)
- Qi Zhang
- School of Life Science and BioEngineering, Beijing University of Technology, Beijing, China (mainland)
| | - Bin Gao
- School of Life Science and BioEngineering, Beijing University of Technology, Beijing, China (mainland)
| | - Chang Yu
- School of Life Science and BioEngineering, Beijing University of Technology, Beijing, China (mainland)
| |
Collapse
|
42
|
Zhu AS, Grande-Allen KJ. Heart valve tissue engineering for valve replacement and disease modeling. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018. [DOI: 10.1016/j.cobme.2017.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
43
|
Nachlas ALY, Li S, Davis ME. Developing a Clinically Relevant Tissue Engineered Heart Valve-A Review of Current Approaches. Adv Healthc Mater 2017; 6. [PMID: 29171921 DOI: 10.1002/adhm.201700918] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/25/2017] [Indexed: 11/08/2022]
Abstract
Tissue engineered heart valves (TEHVs) have the potential to address the shortcomings of current implants through the combination of cells and bioactive biomaterials that promote growth and proper mechanical function in physiological conditions. The ideal TEHV should be anti-thrombogenic, biocompatible, durable, and resistant to calcification, and should exhibit a physiological hemodynamic profile. In addition, TEHVs may possess the capability to integrate and grow with somatic growth, eliminating the need for multiple surgeries children must undergo. Thus, this review assesses clinically available heart valve prostheses, outlines the design criteria for developing a heart valve, and evaluates three types of biomaterials (decellularized, natural, and synthetic) for tissue engineering heart valves. While significant progress has been made in biomaterials and fabrication techniques, a viable tissue engineered heart valve has yet to be translated into a clinical product. Thus, current strategies and future perspectives are also discussed to facilitate the development of new approaches and considerations for heart valve tissue engineering.
Collapse
Affiliation(s)
- Aline L. Y. Nachlas
- Wallace H Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30332 USA
| | - Siyi Li
- Wallace H Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30332 USA
| | - Michael E. Davis
- Wallace H Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30332 USA
- Children's Heart Research & Outcomes (HeRO) Center Children's Healthcare of Atlanta & Emory University Atlanta GA 30322 USA
| |
Collapse
|
44
|
Dahal S, Huang P, Murray BT, Mahler GJ. Endothelial to mesenchymal transformation is induced by altered extracellular matrix in aortic valve endothelial cells. J Biomed Mater Res A 2017; 105:2729-2741. [PMID: 28589644 DOI: 10.1002/jbm.a.36133] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/09/2017] [Accepted: 06/01/2017] [Indexed: 11/10/2022]
Abstract
Alterations in shear stress, mechanical deformation, extracellular matrix (ECM) composition and exposure to inflammatory conditions are known to cause endothelial to mesenchymal transformation (EndMT). This change in endothelial phenotype has only recently been linked to adult pathologies such as cancer progression, organ fibrosis, and calcific aortic valve disease; and its function in adult physiology, especially in response to tissue mechanics, has not been rigorously investigated. EndMT is a response to mechanical and biochemical signals that results in the remodeling of underlying tissues. In diseased aortic valves, glycosaminoglycans (GAGs) are present in the collagen-rich valve fibrosa, and are deposited near calcified nodules. In this study, in vitro models of early and late-stage valve disease were developed by incorporating the GAGs chondroitin sulfate (CS), hyaluronic acid, and dermatan sulfate into 3D collagen hydrogels with or without exposure to TGF-β1 to simulate EndMT in response to microenvironmental changes. High levels of CS induced the highest rate of EndMT and led to the most collagen I and GAG production by mesenchymally transformed cells, which indicates a cell phenotype most likely to promote fibrotic disease. Mesenchymal transformation due to altered ECM was found to depend on cell-ECM bond strength and extracellular signal-regulated protein kinases 1/2 signaling. Determining the environmental conditions that induce and promote EndMT, and the subsequent behavior of mesenchymally transformed cells, will advance understanding on the role of endothelial cells in tissue regeneration or disease progression. © 2017 Wiley Periodicals Inc. J Biomed Mater Res Part A: 105A: 2729-2741, 2017.
Collapse
Affiliation(s)
- Sudip Dahal
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York, USA
| | - Peter Huang
- Department of Mechanical Engineering, Binghamton University, Binghamton, New York, USA
| | - Bruce T Murray
- Department of Mechanical Engineering, Binghamton University, Binghamton, New York, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
45
|
Wang X, Lee J, Ali M, Kim J, Lacerda CMR. Phenotype Transformation of Aortic Valve Interstitial Cells Due to Applied Shear Stresses Within a Microfluidic Chip. Ann Biomed Eng 2017. [DOI: 10.1007/s10439-017-1871-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
46
|
Shen N, Knopf A, Westendorf C, Kraushaar U, Riedl J, Bauer H, Pöschel S, Layland SL, Holeiter M, Knolle S, Brauchle E, Nsair A, Hinderer S, Schenke-Layland K. Steps toward Maturation of Embryonic Stem Cell-Derived Cardiomyocytes by Defined Physical Signals. Stem Cell Reports 2017; 9:122-135. [PMID: 28528699 PMCID: PMC5511039 DOI: 10.1016/j.stemcr.2017.04.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 01/18/2023] Open
Abstract
Cardiovascular disease remains a leading cause of mortality and morbidity worldwide. Embryonic stem cell-derived cardiomyocytes (ESC-CMs) may offer significant advances in creating in vitro cardiac tissues for disease modeling, drug testing, and elucidating developmental processes; however, the induction of ESCs to a more adult-like CM phenotype remains challenging. In this study, we developed a bioreactor system to employ pulsatile flow (1.48 mL/min), cyclic strain (5%), and extended culture time to improve the maturation of murine and human ESC-CMs. Dynamically-cultured ESC-CMs showed an increased expression of cardiac-associated proteins and genes, cardiac ion channel genes, as well as increased SERCA activity and a Raman fingerprint with the presence of maturation-associated peaks similar to primary CMs. We present a bioreactor platform that can serve as a foundation for the development of human-based cardiac in vitro models to verify drug candidates, and facilitates the study of cardiovascular development and disease. Custom-made bioreactor exposes ESC-CMs to defined shear stress and cyclic stretch Physical signals and extended culture significantly improve maturation of ESC-CMs Biochemical fingerprint of dynamically cultured ESC-CMs is similar to primary CMs
Collapse
Affiliation(s)
- Nian Shen
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Anne Knopf
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Claas Westendorf
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany
| | - Udo Kraushaar
- Department of Cell Biology, Electrophysiology, Natural and Medical Sciences Institute, University of Tübingen, Reutlingen 72770, Germany
| | - Julia Riedl
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Hannah Bauer
- Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Simone Pöschel
- Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Shannon Lee Layland
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Monika Holeiter
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Stefan Knolle
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Cell Biology, Electrophysiology, Natural and Medical Sciences Institute, University of Tübingen, Reutlingen 72770, Germany
| | - Eva Brauchle
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Ali Nsair
- Department of Medicine/Cardiology, Cardiovascular Research Laboratories (CVRL), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA; Broad Stem Cell Research Center, David School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Svenja Hinderer
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany
| | - Katja Schenke-Layland
- Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany; Department of Women's Health, Research Institute of Women's Health, University Hospital of the Eberhard Karls University Tübingen, Tübingen 72076, Germany; Department of Medicine/Cardiology, Cardiovascular Research Laboratories (CVRL), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| |
Collapse
|
47
|
Wu B, Wang Y, Xiao F, Butcher JT, Yutzey KE, Zhou B. Developmental Mechanisms of Aortic Valve Malformation and Disease. Annu Rev Physiol 2017; 79:21-41. [DOI: 10.1146/annurev-physiol-022516-034001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Bingruo Wu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
| | - Yidong Wang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
| | - Feng Xiao
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029 China
| | - Jonathan T. Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853;
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio 45229;
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029 China
| |
Collapse
|
48
|
Liu X, Xu Z. Osteogenesis in calcified aortic valve disease: From histopathological observation towards molecular understanding. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 122:156-161. [DOI: 10.1016/j.pbiomolbio.2016.02.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/23/2016] [Accepted: 02/25/2016] [Indexed: 12/14/2022]
|
49
|
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
|
50
|
Maeda K, Ma X, Chalajour F, Hanley FL, Riemer RK. Critical Role of Coaptive Strain in Aortic Valve Leaflet Homeostasis: Use of a Novel Flow Culture Bioreactor to Explore Heart Valve Mechanobiology. J Am Heart Assoc 2016; 5:JAHA.116.003506. [PMID: 27464792 PMCID: PMC5015277 DOI: 10.1161/jaha.116.003506] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Aortic valve (AV) disease presents critical situations requiring surgery in over 2% of the US population and is increasingly the reason for cardiac surgery. Throughout the AV cycle, mechanical forces of multiple types and varying intensities are exerted on valve leaflets. The mechanisms whereby forces regulate leaflet homeostasis are incompletely understood. We used a novel flow bioreactor culture to investigate alteration of AV opening or closure on leaflet genes. Methods and Results Culture of rat AV was conducted in a flow bioreactor for 7 days at 37°C under conditions approximating the normal stroke volume. Three force condition groups were compared: Cycling (n=8); always open (Open; n=3); or always closed (Closed; n=5). From each culture, AV leaflets were pooled by force condition and RNA expression evaluated using microarrays. Hierarchical clustering of 16 transcriptome data sets from the 3 groups revealed only 2 patterns of gene expression: Cycling and Closed groups clustered together, whereas Open AV were different (P<0.05). Sustained AV opening induced marked changes in expression (202 transcripts >2‐fold; P<0.05), whereas Closed AV exhibited similar expression pattern as Cycling (no transcripts >2‐fold; P<0.05). Comparison with human sclerotic and calcific AV transcriptomes demonstrated high concordance of >40 Open group genes with progression toward disease. Conclusions Failure of AV to close initiates an extensive response characterized by expression changes common to progression to calcific aortic valve disease. AV coaptation, whether phasic or chronic, preserved phenotypic gene expression. These results demonstrate, for the first time, that coaptation of valve leaflets is a fundamentally important biomechanical cue driving homeostasis.
Collapse
Affiliation(s)
- Katsuhide Maeda
- Pediatric Cardiac Surgery Division, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Falk Cardiovascular Research Center, Stanford, CA
| | - Xiaoyuan Ma
- Pediatric Cardiac Surgery Division, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Falk Cardiovascular Research Center, Stanford, CA
| | - Fariba Chalajour
- Pediatric Cardiac Surgery Division, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Falk Cardiovascular Research Center, Stanford, CA
| | - Frank L Hanley
- Pediatric Cardiac Surgery Division, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Falk Cardiovascular Research Center, Stanford, CA
| | - R Kirk Riemer
- Pediatric Cardiac Surgery Division, Department of Cardiothoracic Surgery, Stanford University School of Medicine, Falk Cardiovascular Research Center, Stanford, CA
| |
Collapse
|