1
|
Huang Y, Wang C, Zhou T, Xie F, Liu Z, Xu H, Liu M, Wang S, Li L, Chi Q, Shi J, Dong N, Xu K. Lumican promotes calcific aortic valve disease through H3 histone lactylation. Eur Heart J 2024; 45:3871-3885. [PMID: 38976370 DOI: 10.1093/eurheartj/ehae407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/06/2023] [Accepted: 06/18/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND AND AIMS Valve interstitial cells (VICs) undergo a transition to intermediate state cells before ultimately transforming into the osteogenic cell population, which is a pivotal cellular process in calcific aortic valve disease (CAVD). Herein, this study successfully delineated the stages of VIC osteogenic transformation and elucidated a novel key regulatory role of lumican (LUM) in this process. METHODS Single-cell RNA-sequencing (scRNA-seq) from nine human aortic valves was used to characterize the pathological switch process and identify key regulatory factors. The in vitro, ex vivo, in vivo, and double knockout mice were constructed to further unravel the calcification-promoting effect of LUM. Moreover, the multi-omic approaches were employed to analyse the molecular mechanism of LUM in CAVD. RESULTS ScRNA-seq successfully delineated the process of VIC pathological transformation and highlighted the significance of LUM as a novel molecule in this process. The pro-calcification role of LUM is confirmed on the in vitro, ex vivo, in vivo level, and ApoE-/-//LUM-/- double knockout mice. The LUM induces osteogenesis in VICs via activation of inflammatory pathways and augmentation of cellular glycolysis, resulting in the accumulation of lactate. Subsequent investigation has unveiled a novel LUM driving histone modification, lactylation, which plays a role in facilitating valve calcification. More importantly, this study has identified two specific sites of histone lactylation, namely, H3K14la and H3K9la, which have been found to facilitate the process of calcification. The confirmation of these modification sites' association with the expression of calcific genes Runx2 and BMP2 has been achieved through ChIP-PCR analysis. CONCLUSIONS The study presents novel findings, being the first to establish the involvement of lumican in mediating H3 histone lactylation, thus facilitating the development of aortic valve calcification. Consequently, lumican would be a promising therapeutic target for intervention in the treatment of CAVD.
Collapse
Affiliation(s)
- Yuming Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunli Wang
- Hubei Shizhen Laboratory, Wuhan 430065, China
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Tingwen Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fei Xie
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zongtao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haiying Xu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ming Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shunshun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Lanqing Li
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Qingjia Chi
- Department of Engineering Structure and Mechanics, School of Science, Wuhan University of Technology, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, 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
| | - Kang Xu
- Hubei Shizhen Laboratory, Wuhan 430065, China
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| |
Collapse
|
2
|
Mach F, Miteva K. Window of opportunity for developing effective medical intervention for calcific aortic valve disease. Eur Heart J 2024; 45:3886-3888. [PMID: 39132886 DOI: 10.1093/eurheartj/ehae426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/13/2024] Open
Affiliation(s)
- François Mach
- Division of Cardiology, Geneva University Hospital & Faculty of Medicine, Geneva, Switzerland
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Research, Department of Medicine, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland
| |
Collapse
|
3
|
Poznyak A, Kashirskikh D, Postnov A, Popov M, Sukhorukov V, Orekhov A. Sialic acid as the potential link between lipid metabolism and inflammation in the pathogenesis of atherosclerosis. Braz J Med Biol Res 2023; 56:e12972. [PMID: 38088673 PMCID: PMC10712282 DOI: 10.1590/1414-431x2023e12972] [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: 07/22/2023] [Accepted: 10/28/2023] [Indexed: 12/18/2023] Open
Abstract
In the modern world, cardiovascular diseases have a special place among the most common causes of death. Naturally, this widespread problem cannot escape the attention of scientists and researchers. One of the main conditions preceding the development of fatal cardiovascular diseases is atherosclerosis. Despite extensive research into its pathogenesis and possible prevention and treatment strategies, many gaps remain in our understanding of this disease. For example, the concept of multiple low-density lipoprotein modifications was recently stated, in which desialylation is of special importance. Apart from this, sialic acids are known to be important contributors to processes such as endothelial dysfunction and inflammation, which in turn are major components of atherogenesis. In this review, we have collected information on sialic acid metabolism, analyzed various aspects of its implication in atherosclerosis at different stages, and provided an overview of the role of particular groups of enzymes responsible for sialic acid metabolism in the context of atherosclerosis.
Collapse
Affiliation(s)
- A.V. Poznyak
- Institute for Atherosclerosis Research, Moscow, Russia
| | | | - A.Y. Postnov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | - M.A. Popov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), Moscow, Russia
| | - V.N. Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | - A.N. Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| |
Collapse
|
4
|
Poznyak AV, Orekhova VA, Sukhorukov VN, Khotina VA, Popov MA, Orekhov AN. Atheroprotective Aspects of Heat Shock Proteins. Int J Mol Sci 2023; 24:11750. [PMID: 37511509 PMCID: PMC10380699 DOI: 10.3390/ijms241411750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Atherosclerosis is a major global health problem. Being a harbinger of a large number of cardiovascular diseases, it ultimately leads to morbidity and mortality. At the same time, effective measures for the prevention and treatment of atherosclerosis have not been developed, to date. All available therapeutic options have a number of limitations. To understand the mechanisms behind the triggering and development of atherosclerosis, a deeper understanding of molecular interactions is needed. Heat shock proteins are important for the normal functioning of cells, actively helping cells adapt to gradual changes in the environment and survive in deadly conditions. Moreover, multiple HSP families play various roles in the progression of cardiovascular disorders. Some heat shock proteins have been shown to have antiatherosclerotic effects, while the role of others remains unclear. In this review, we considered certain aspects of the antiatherosclerotic activity of a number of heat shock proteins.
Collapse
Affiliation(s)
- Anastasia V Poznyak
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Varvara A Orekhova
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Vasily N Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Victoria A Khotina
- Institute of General Pathology and Pathophysiology, 8, Baltiyskaya St., 125315 Moscow, Russia
| | - Mikhail A Popov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), 61/2, Shchepkin St., 129110 Moscow, Russia
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| |
Collapse
|
5
|
Mamazhakypov A, Maripov A, Sarybaev AS, Schermuly RT, Sydykov A. Osteopontin in Pulmonary Hypertension. Biomedicines 2023; 11:biomedicines11051385. [PMID: 37239056 DOI: 10.3390/biomedicines11051385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Pulmonary hypertension (PH) is a pathological condition with multifactorial etiology, which is characterized by elevated pulmonary arterial pressure and pulmonary vascular remodeling. The underlying pathogenetic mechanisms remain poorly understood. Accumulating clinical evidence suggests that circulating osteopontin may serve as a biomarker of PH progression, severity, and prognosis, as well as an indicator of maladaptive right ventricular remodeling and dysfunction. Moreover, preclinical studies in rodent models have implicated osteopontin in PH pathogenesis. Osteopontin modulates a plethora of cellular processes within the pulmonary vasculature, including cell proliferation, migration, apoptosis, extracellular matrix synthesis, and inflammation via binding to various receptors such as integrins and CD44. In this article, we provide a comprehensive overview of the current understanding of osteopontin regulation and its impact on pulmonary vascular remodeling, as well as consider research issues required for the development of therapeutics targeting osteopontin as a potential strategy for the management of PH.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Abdirashit Maripov
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Akpay S Sarybaev
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Akylbek Sydykov
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| |
Collapse
|
6
|
Calcific aortic valve disease: mechanisms, prevention and treatment. Nat Rev Cardiol 2023:10.1038/s41569-023-00845-7. [PMID: 36829083 DOI: 10.1038/s41569-023-00845-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/01/2023] [Indexed: 02/26/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most common disorder affecting heart valves and is characterized by thickening, fibrosis and mineralization of the aortic valve leaflets. Analyses of surgically explanted aortic valve leaflets have shown that dystrophic mineralization and osteogenic transition of valve interstitial cells co-occur with neovascularization, microhaemorrhage and abnormal production of extracellular matrix. Age and congenital bicuspid aortic valve morphology are important and unalterable risk factors for CAVD, whereas additional risk is conferred by elevated blood pressure and plasma lipoprotein(a) levels and the presence of obesity and diabetes mellitus, which are modifiable factors. Genetic and molecular studies have identified that the NOTCH, WNT-β-catenin and myocardin signalling pathways are involved in the control and commitment of valvular cells to a fibrocalcific lineage. Complex interactions between valve endothelial and interstitial cells and immune cells promote the remodelling of aortic valve leaflets and the development of CAVD. Although no medical therapy is effective for reducing or preventing the progression of CAVD, studies have started to identify actionable targets.
Collapse
|
7
|
The Role of Macrophages in the Pathogenesis of Atherosclerosis. Cells 2023; 12:cells12040522. [PMID: 36831189 PMCID: PMC9954519 DOI: 10.3390/cells12040522] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
A wide variety of cell populations, including both immune and endothelial cells, participate in the pathogenesis of atherosclerosis. Among these groups, macrophages deserve special attention because different populations of them can have completely different effects on atherogenesis and inflammation in atherosclerosis. In the current review, the significance of different phenotypes of macrophages in the progression or regression of atherosclerosis will be considered, including their ability to become the foam cells and the consequences of this event, as well as their ability to create a pro-inflammatory or anti-inflammatory medium at the site of atherosclerotic lesions as a result of cytokine production. In addition, several therapeutic strategies directed to the modulation of macrophage activity, which can serve as useful ideas for future drug developments, will be considered.
Collapse
|
8
|
Poznyak AV, Sukhorukov VN, Surkova R, Orekhov NA, Orekhov AN. Glycation of LDL: AGEs, impact on lipoprotein function, and involvement in atherosclerosis. Front Cardiovasc Med 2023; 10:1094188. [PMID: 36760567 PMCID: PMC9904536 DOI: 10.3389/fcvm.2023.1094188] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023] Open
Abstract
Atherosclerosis is a complex disease, and there are many factors that influence its development and the course of the disease. A deep understanding of the pathological mechanisms underlying atherogenesis is needed to develop optimal therapeutic strategies and treatments. In this review, we have focused on low density lipoproteins. According to multiple studies, their atherogenic properties are associated with multiple modifications of lipid particles. One of these modifications is Glycation. We considered aspects related to the formation of modified particles, as well as the influence of modification on their functioning. We paid special attention to atherogenicity and the role of glycated low-density lipoprotein (LDL) in atherosclerosis.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Moscow, Russia,*Correspondence: Anastasia V. Poznyak,
| | - Vasily N. Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Raisa Surkova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Nikolay A. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Moscow, Russia,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
9
|
Ground M, Park YE, Waqanivavalagi S, Callon K, Walker R, Milsom P, Cornish J. Generating robust human valvular interstitial cell cultures: Protocol and considerations. J Mol Cell Cardiol 2022; 173:118-126. [PMID: 36327771 DOI: 10.1016/j.yjmcc.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/19/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Research in heart valve biology is a growing field that has yet to elucidate the fundamentals of valve disease. Human valvular interstitial cells (hVICs) are the best option for studying the cellular mechanisms behind valvular pathologies. However, there is a wide range of isolation procedures for these cells published in the literature. To what extent various isolation methods, patient pathologies, and seeding densities influence the behaviour of hVICs remains unclear. Here, we present an optimised method of hVIC isolation from diseased human valves donated at the time of surgery. We show that two rounds of 1000 U/mL collagenase digestion for not >2 h results in a phenotypically stable cell culture with a near complete absence of endothelial cell contamination. We also suggest that cells should be seeded at 10,000 cells/cm2 for experimentation. We found that patient pathology does not affect the success of the isolation procedure, and that instead, successful cultures are predicted by ensuring >500 mg valve tissue as starting material.
Collapse
Affiliation(s)
- Marcus Ground
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.
| | - Young Eun Park
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, 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
| | - 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
|
10
|
Targeting Mitochondrial Dynamics Proteins for the Development of Therapies for Cardiovascular Diseases. Int J Mol Sci 2022; 23:ijms232314741. [PMID: 36499064 PMCID: PMC9736032 DOI: 10.3390/ijms232314741] [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: 10/28/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular diseases are one of the leading causes of death worldwide. The identification of new pathogenetic targets contributes to more efficient development of new types of drugs for the treatment of cardiovascular diseases. This review highlights the problem of mitochondrial dynamics disorders, in the context of cardiovascular diseases. A change in the normal function of mitochondrial dynamics proteins is one of the reasons for the development of the pathological state of cardiomyocytes. Based on this, therapeutic targeting of these proteins may be a promising strategy in the development of cardiac drugs. Here we will consider changes for each process of mitochondrial dynamics in cardiovascular diseases: fission and fusion of mitochondria, mitophagy, mitochondrial transport and biogenesis, and also analyze the prospects of the considered protein targets based on existing drug developments.
Collapse
|
11
|
Ozawa K, Muller MA, Varlamov O, Hagen MW, Packwood W, Morgan TK, Xie A, López CS, Chung D, Chen J, López JA, Lindner JR. Reduced Proteolytic Cleavage of von Willebrand Factor Leads to Aortic Valve Stenosis and Load-Dependent Ventricular Remodeling. JACC Basic Transl Sci 2022; 7:642-655. [PMID: 35958695 PMCID: PMC9357566 DOI: 10.1016/j.jacbts.2022.02.021] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/03/2022] [Accepted: 02/17/2022] [Indexed: 11/28/2022]
Abstract
We hypothesized that excess endothelial-associated von Willebrand factor (vWF) and secondary platelet adhesion contribute to aortic valve stenosis (AS). We studied hyperlipidemic mice lacking ADAMTS13 (LDLR -/- AD13 -/- ), which cleaves endothelial-associated vWF multimers. On echocardiography and molecular imaging, LDLR -/- AD13 -/- compared with control strains had increased aortic endothelial vWF and platelet adhesion and developed hemodynamically significant AS, arterial stiffening, high valvulo-aortic impedance, and secondary load-dependent reduction in LV systolic function. Histology revealed leaflet thickening and calcification with valve interstitial cell myofibroblastic and osteogenic transformation, and evidence for TGFβ1 pathway activation. We conclude that valve leaflet endothelial vWF-platelet interactions promote AS through juxtacrine platelet signaling.
Collapse
Affiliation(s)
- Koya Ozawa
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Matthew A. Muller
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Oleg Varlamov
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Matthew W. Hagen
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Terry K. Morgan
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Aris Xie
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Claudia S. López
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | | | | | | | - Jonathan R. Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
- Address for correspondence: Dr Jonathan R. Lindner, Cardiovascular Division, UHN-62, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA. @JLindnerMD
| |
Collapse
|
12
|
Bogdanova M, Zabirnyk A, Malashicheva A, Semenova D, Kvitting JPE, Kaljusto ML, Perez MDM, Kostareva A, Stensløkken KO, Sullivan GJ, Rutkovskiy A, Vaage J. Models and Techniques to Study Aortic Valve Calcification in Vitro, ex Vivo and in Vivo. An Overview. Front Pharmacol 2022; 13:835825. [PMID: 35721220 PMCID: PMC9203042 DOI: 10.3389/fphar.2022.835825] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Aortic valve stenosis secondary to aortic valve calcification is the most common valve disease in the Western world. Calcification is a result of pathological proliferation and osteogenic differentiation of resident valve interstitial cells. To develop non-surgical treatments, the molecular and cellular mechanisms of pathological calcification must be revealed. In the current overview, we present methods for evaluation of calcification in different ex vivo, in vitro and in vivo situations including imaging in patients. The latter include echocardiography, scanning with computed tomography and magnetic resonance imaging. Particular emphasis is on translational studies of calcific aortic valve stenosis with a special focus on cell culture using human primary cell cultures. Such models are widely used and suitable for screening of drugs against calcification. Animal models are presented, but there is no animal model that faithfully mimics human calcific aortic valve disease. A model of experimentally induced calcification in whole porcine aortic valve leaflets ex vivo is also included. Finally, miscellaneous methods and aspects of aortic valve calcification, such as, for instance, biomarkers are presented.
Collapse
Affiliation(s)
- Maria Bogdanova
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Arsenii Zabirnyk
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Daria Semenova
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | | | - Mari-Liis Kaljusto
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Woman and Children Health, Karolinska Institute, Stockholm, Sweden
| | - Kåre-Olav Stensløkken
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Gareth J Sullivan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Institute of Immunology, Oslo University Hospital, Oslo, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Arkady Rutkovskiy
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pulmonary Diseases, Oslo University Hospital, Oslo, Norway
| | - Jarle Vaage
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
13
|
Huang K, Wu L, Gao Y, Li Q, Wu H, Liu X, Han L. Transcriptome Sequencing Data Reveal LncRNA-miRNA-mRNA Regulatory Network in Calcified Aortic Valve Disease. Front Cardiovasc Med 2022; 9:886995. [PMID: 35722091 PMCID: PMC9204424 DOI: 10.3389/fcvm.2022.886995] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundCalcified aortic valve disease (CAVD) is one of the most common valvular heart diseases in the elderly population. However, no effective medical treatments have been found to interfere with the progression of CAVD, and specific molecular mechanisms of CAVD remain unclear.Materials and MethodsTranscriptome sequencing data of GSE55492 and GSE148219 were downloaded from the European Nucleotide Archive, and the microarray dataset, GSE12644 was acquired from the Gene Expression Omnibus database. Software, including FastQC, HISAT2, samtools, and featureCounts was applied to generate the read count matrix. The “Limma” package in R was utilized to analyze differentially expressed genes (DEGs). Thereafter, weighted gene co-expression network analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and the protein-protein interaction (PPI) network were used to identify hub genes associated with CAVD, which were further validated by receiver operating characteristic curve (ROC) analysis using GSE12644. The long non-coding RNA (LncRNA)-mediated regulatory network was established based on the differentially expressed LncRNAs and hub genes, which were detected using quantitative real-time PCR (qRT-PCR) in clinical samples and valve interstitial cells. Moreover, CIBERSORT was used to calculate the expression distribution of immune cell infiltration in CAVD.ResultsA total of 126 DEGs were included in the PPI network. PI3K-Akt signaling pathway, ECM-receptor interaction, hematopoietic cell lineage, cell adhesion molecules, and focal adhesion were the most enriched pathways revealed by KEGG. Four LncRNAs, including TRHDE-AS1, LINC00092, LINC01094, and LINC00702 were considered the differentially expressed LncRNA. SPP1, TREM1, GPM6A, CCL19, CR1, NCAM1, CNTN1, TLR8, SDC1, and COL6A6 were the 10 hub genes identified to be associated with CAVD. Moreover, the calcified aortic valve samples had a greater level of Tregs, naïve B cells, and M0 macrophages than the noncalcified ones, whereas CAVD samples had a lower M2 macrophage expression compared to the noncalcified valve tissues.ConclusionThe current study identified SPP1, TREM1, TLR8, SDC1, GPM6A, and CNTN1 as hub genes that could potentially be associated with CAVD. The LINC00702–miR-181b-5p–SPP1 axis might participate in the development of CAVD. Additionally, M2 macrophages, Tregs, naïve B cells, and M0 macrophages might possibly play a role in the initiation of CAVD.
Collapse
|
14
|
Moschetta D, Di Maria E, Valerio V, Massaiu I, Bozzi M, Songia P, D’alessandra Y, Myasoedova VA, Poggio P. Purinergic Receptor P2Y2 Stimulation Averts Aortic Valve Interstitial Cell Calcification and Myofibroblastic Activation. Biomedicines 2022; 10:biomedicines10020457. [PMID: 35203666 PMCID: PMC8962345 DOI: 10.3390/biomedicines10020457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
Rationale—Calcific aortic valve stenosis (CAVS) is a pathological condition of the aortic valve with a prevalence of 3% in the general population. It is characterized by massive rearrangement of the extracellular matrix, mostly due to the accumulation of fibro-calcific deposits driven by valve interstitial cells (VIC), and no pharmacological treatment is currently available. The aim of this study was to evaluate the effects of P2Y2 receptor (P2RY2) activation on fibro-calcific remodeling of CAVS. Methods—We employed human primary VICs isolated from CAVS leaflets treated with 2-thiouridine-5′-triphosphate (2ThioUTP, 10 µM), an agonist of P2RY2. The calcification was induced by inorganic phosphate (2 mM) and ascorbic acid (50 µg/mL) for 7 or 14 days, while the 2ThioUTP was administered starting from the seventh day. 2ThioUTP was chronically administered for 5 days to evaluate myofibroblastic activation. Results—P2RY2 activation, under continuous or interrupted pro-calcific stimuli, led to a significant inhibition of VIC calcification potential (p < 0.01). Moreover, 2ThioUTP treatment was able to significantly reduce pro-fibrotic gene expression (p < 0.05), as well as that of protein α-smooth muscle actin (p = 0.004). Conclusions—Our data suggest that P2RY2 activation should be further investigated as a pharmacological target for the prevention of CAVS progression, acting on both calcification and myofibroblastic activation.
Collapse
Affiliation(s)
- Donato Moschetta
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Enrico Di Maria
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
| | - Vincenza Valerio
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
| | - Ilaria Massaiu
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Michele Bozzi
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
| | - Paola Songia
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
| | - Yuri D’alessandra
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
| | - Veronika A. Myasoedova
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
| | - Paolo Poggio
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (D.M.); (E.D.M.); (V.V.); (I.M.); (M.B.); (P.S.); (Y.D.); (V.A.M.)
- Correspondence: ; Tel.: +39-02-5800-2853
| |
Collapse
|
15
|
Zhang F, Cheng N, Han Y, Zhang C, Zhang H. miRNA Expression Profiling Uncovers a Role of miR-139-5p in Regulating the Calcification of Human Aortic Valve Interstitial Cells. Front Genet 2021; 12:722564. [PMID: 34745206 PMCID: PMC8569802 DOI: 10.3389/fgene.2021.722564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/27/2021] [Indexed: 01/02/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common structural heart disease, and the morbidity is increased with elderly population. Several microRNAs (miRNAs) have been identified to play crucial roles in CAVD, and numerous miRNAs are still waiting to be explored. In this study, the miRNA expression signature in CAVD was analyzed unbiasedly by miRNA-sequencing, and we found that, compared with the normal control valves, 152 miRNAs were upregulated and 186 miRNAs were downregulated in calcified aortic valves. The functions of these differentially expressed miRNAs were associated with cell differentiation, apoptosis, adhesion and immune response processes. Among downregulated miRNAs, the expression level of miR-139-5p was negatively correlated with the osteogenic gene RUNX2, and miR-139-5p was also downregulated during the osteogenic differentiation of primary human aortic valve interstitial cells (VICs). Subsequent functional studies revealed that miR-139-5p overexpression inhibited the osteogenic differentiation of VICs by negatively modulating the expression of pro-osteogenic gene FZD4 and CTNNB1. In conclusion, these results suggest that miR-139-5p plays an important role in osteogenic differentiation of VICs via the Wnt/β-Catenin pathway, which may further provide a new therapeutic target for CAVD.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Naixuan Cheng
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Vascular Diseases, Beijing, China
| | - Yingchun Han
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Vascular Diseases, Beijing, China
| | - Congcong Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Vascular Diseases, Beijing, China
| | - Haibo Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Jiang R, Tran M, Lönnerdal B. Recombinant Bovine and Human Osteopontin Generated by Chlamydomonas reinhardtii Exhibit Bioactivities Similar to Bovine Milk Osteopontin When Assessed in Mouse Pups Fed Osteopontin-Deficient Milk. Mol Nutr Food Res 2021; 65:e2000644. [PMID: 34050612 DOI: 10.1002/mnfr.202000644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 05/11/2021] [Indexed: 11/09/2022]
Abstract
SCOPE Osteopontin (OPN), a highly phosphorylated and glycosylated protein, is present in most body fluids, including milk. OPN appears at a high concentration in human milk (130-180 mg L-1 ), but not bovine milk (≈18 mg mL-1 ). It is previously shown that milk OPN is involved in various biological processes and therefore may be a valuable infant formula additive. METHODS AND RESULTS In the present study, recombinant bovine OPN (rbOPN) and recombinant human OPN (rhOPN) are generated in a Chlamydomonas reinhardtii (C. reinhardtii) algal expression system. The rbOPN and rhOPN are phosphorylated but not glycosylated. To assess the bioactivities of rbOPN and rhOPN and compare their bioactivities to those of bovine milk OPN (bmOPN), wild-type (WT) mouse pups nursed by OPN knock-out (KO) dams are orally fed bmOPN, rbOPN, and rhOPN daily from postnatal days 1-21 (P1-21). Effects of these OPNs on development of the brain, intestine, and immune function are evaluated. The results show that rbOPN and rhOPN exhibit effects similar to those of bmOPN as well as mouse milk OPN on stimulating proliferation of the small intestine, increasing brain myelination and cognitive development, and enhancing development of immune function. CONCLUSION rbOPN and rhOPN are likely to provide beneficial bioactivities when added to infant diets.
Collapse
Affiliation(s)
- Rulan Jiang
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Miller Tran
- Triton Algae Innovations, San Diego, CA, 92121, USA
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| |
Collapse
|
17
|
Howsmon DP, Sacks MS. On Valve Interstitial Cell Signaling: The Link Between Multiscale Mechanics and Mechanobiology. Cardiovasc Eng Technol 2021; 12:15-27. [PMID: 33527256 PMCID: PMC11046423 DOI: 10.1007/s13239-020-00509-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/05/2020] [Indexed: 01/02/2023]
Abstract
Heart valves function in one of the most mechanically demanding environments in the body to ensure unidirectional blood flow. The resident valve interstitial cells respond to this mechanical environment and maintain the structure and integrity of the heart valve tissues to preserve homeostasis. While the mechanics of organ-tissue-cell heart valve function has progressed, the intracellular signaling network downstream of mechanical stimuli has not been fully elucidated. This has hindered efforts to both understand heart valve mechanobiology and rationally identify drug targets for treating valve disease. In the present work, we review the current literature on VIC mechanobiology and then propose mechanistic mathematical modeling of the mechanically-stimulated VIC signaling response to comprehend the coupling between VIC mechanobiology and valve mechanics.
Collapse
Affiliation(s)
- Daniel P Howsmon
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Michael S Sacks
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
18
|
Extracellular Matrix in Calcific Aortic Valve Disease: Architecture, Dynamic and Perspectives. Int J Mol Sci 2021; 22:ijms22020913. [PMID: 33477599 PMCID: PMC7831300 DOI: 10.3390/ijms22020913] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/18/2022] Open
Abstract
Calcific Aortic Valve Disease (CAVD) is the most common valvular heart disease in developed countries and in the ageing population. It is strongly correlated to median age, affecting up to 13% of the population over the age of 65. Pathophysiological analysis indicates CAVD as a result of an active and degenerative disease, starting with sclerosis and chronic inflammation and then leaflet calcification, which ultimately can account for aortic stenosis. Although CAVD has been firstly recognized as a passive event mostly resulting from a degenerative aging process, much evidences suggests that calcification arises from different active processes, involving both aortic valve-resident cells (valve endothelial cells, valve interstitial cells, mesenchymal stem cells, innate immunity cells) and circulating cells (circulating mesenchymal cells, immunity cells). Moreover, a role for the cell-derived "matrix vesicles" and extracellular matrix (ECM) components has also been recognized. The aim of this work is to review the cellular and molecular alterations occurring in aortic valve during CAVD pathogenesis, focusing on the role of ECM in the natural course of the disease.
Collapse
|
19
|
Bourgeois R, Girard A, Perrot N, Guertin J, Mitchell PL, Couture C, Gotti C, Bourassa S, Poggio P, Mass E, Capoulade R, Scipione CA, Després AA, Couture P, Droit A, Pibarot P, Boffa MB, Thériault S, Koschinsky ML, Mathieu P, Arsenault BJ. A Comparative Analysis of the Lipoprotein(a) and Low-Density Lipoprotein Proteomic Profiles Combining Mass Spectrometry and Mendelian Randomization. CJC Open 2020; 3:450-459. [PMID: 34027348 PMCID: PMC8129481 DOI: 10.1016/j.cjco.2020.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 11/28/2020] [Indexed: 12/30/2022] Open
Abstract
Background Lipoprotein(a) (Lp[a]), which consists of a low-density lipoprotein (LDL) bound to apolipoprotein(a), is one of the strongest genetic risk factors for atherosclerotic cardiovascular diseases. Few studies have performed hypothesis-free direct comparisons of the Lp(a) and the LDL proteomes. Our objectives were to compare the Lp(a) and the LDL proteomic profiles and to evaluate the effect of lifelong exposure to elevated Lp(a) or LDL cholesterol levels on the plasma proteomic profile. Methods We performed a label-free analysis of the Lp(a) and LDL proteomic profiles of healthy volunteers in a discovery (n = 6) and a replication (n = 9) phase. We performed inverse variance weighted Mendelian randomization to document the effect of lifelong exposure to elevated Lp(a) or LDL cholesterol levels on the plasma proteomic profile of participants of the INTERVAL study. Results We identified 15 proteins that were more abundant on Lp(a) compared with LDL (serping1, pi16, itih1, itih2, itih3, pon1, podxl, cd44, cp, ptprg, vtn, pcsk9, igfals, vcam1, and ttr). We found no proteins that were more abundant on LDL compared with Lp(a). After correction for multiple testing, lifelong exposure to elevated LDL cholesterol levels was associated with the variation of 18 plasma proteins whereas Lp(a) did not appear to influence the plasma proteome. Conclusions Results of this study highlight marked differences in the proteome of Lp(a) and LDL as well as in the effect of lifelong exposure to elevated LDL cholesterol or Lp(a) on the plasma proteomic profile.
Collapse
Affiliation(s)
- Raphaëlle Bourgeois
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Arnaud Girard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Nicolas Perrot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Jakie Guertin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Patricia L Mitchell
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada
| | - Christian Couture
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada
| | - Clarisse Gotti
- Proteomics platform of the CHU de Québec, Quebec, Canada
| | | | | | - Elvira Mass
- University of Bonn, Developmental Biology of the Immune System, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Romain Capoulade
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Corey A Scipione
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Audrey-Anne Després
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Patrick Couture
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada.,Centre de recherche du CHU de Québec, Quebec, Canada
| | - Arnaud Droit
- Proteomics platform of the CHU de Québec, Quebec, Canada.,Centre de recherche du CHU de Québec, Quebec, Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Michael B Boffa
- Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| |
Collapse
|
20
|
Are there any subclinical myocardial dysfunctions in subjects with aortic valve sclerosis? A 3D-speckle tracking echocardiography study. Int J Cardiovasc Imaging 2020; 37:207-213. [PMID: 32888107 DOI: 10.1007/s10554-020-01977-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
Aortic valve sclerosis (AVS) is defined as calcified and thickened aortic leaflets without restriction of leaflet motion. We have not found any studies that previously assessed the effect of AVS on myocardial functions with three dimensional-speckle tracking echocardiography (3D-STE). Therefore, we aimed to identify any early changes in left atrial (LA) myocardial dynamics and/or left ventricular (LV) systolic functions in patients with AVS using 3D-STE. Seventy-five patients with AVS and 80 age- and gender-matched controls were enrolled into the study. The baseline clinical characteristics of the study patients were recorded. Conventional 2D echocardiographic and 3D-STE analyses were performed. The LV-global longitudinal strain (LV-GLS) and LV-global circumferential strain (LV-GCS) were significantly decreased in the AVS (+) group than in the control group (p < 0.001 and p = 0.013, respectively). In multivariate logistic regression analysis; LV-GLS (p < 0.001, odds ratio (OR) = 3.16, 95% confidence interval (CI) 1.42-5.63) and Triglyceride (TG) (p = 0.033, OR = 1.29, 95% CI 1.11-1.72) were found to be independent predictors of AVS. ROC analysis was performed to find out the ideal LV-GLS cut-off value for predicting the AVS. A LV-GLS value of > - 18 has 85.8% sensitivity, 67.5% specificity for the prediction of the AVS. Our results support that subjects with AVS may have subclinical LV deformation abnormalities even though they have not LV pressure overload. According to our findings, patients with AVS should be investigated in terms of atherosclerotic risk factors, their dysmetabolic status should be evaluated and closely followed up for their progression to calcific aortic stenosis.
Collapse
|
21
|
Myasoedova VA, Di Minno A, Songia P, Massaiu I, Alfieri V, Valerio V, Moschetta D, Andreini D, Alamanni F, Pepi M, Trabattoni D, Poggio P. Sex-specific differences in age-related aortic valve calcium load: A systematic review and meta-analysis. Ageing Res Rev 2020; 61:101077. [PMID: 32334093 DOI: 10.1016/j.arr.2020.101077] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 11/28/2022]
Abstract
Aging of the aortic valve, characterized by leaflet thickening and loss of extensibility, leads to progressive changes in valve function. These age-related mechanisms have not been evaluated yet in sex-specific calcific aortic valve stenosis (CAVS) onset and progression. Recent studies reported the association between high aortic valve calcification (AVC) load and male gender in patients with CAVS while women present faster progression than men. To evaluate these age- and sex-specific differences, we performed a systematic review and meta-analysis with meta-regression. A systematic search related to AVC measured by computed tomography and gender-specific differences was conducted according to PRISMA guidelines. Seven studies, enrolling 1859 men and 1055 women, were included in the quantitative synthesis. We found a significant difference between men and women both in AVC load and density. AVC load mean difference (MD), between men and women, was 1131 ± 243 AU (p < 0.0001; I2: 96.5 %, p < 0.001), while AVC density MD was 159 ± 20 AU/cm2 (p < 0.0001) without heterogeneity among the studies (I2: 23.5, p = 0.3). Meta-regression analyses showed that AVC load MD positively correlated with age and other cardiovascular risk factors such as diabetes, hypertension, and coronary artery disease presence. Our meta-analysis shows a significant association of incremental AVC load with male gender, regardless of the individual anatomical characteristics and the cardiovascular risk factors. Further studies are needed: i) to clarify if there are different sex-related pathophysiological processes driving the development and the progression of age-related CAVS, and ii) to determine if a sex-specific therapeutic strategy should be applied for CAVS treatment and/or prevention.
Collapse
Affiliation(s)
| | - Alessandro Di Minno
- Università deli Studi di Napoli Federico II, Dipartimento di Farmacia, Napoli, Italy
| | | | | | | | - Vincenza Valerio
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Università deli Studi di Napoli Federico II, Dipartimento di Medicina Clinica e Chirurgia, Napoli, Italy
| | - Donato Moschetta
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Università degli Studi di Milano, Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy
| | - Daniele Andreini
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Università degli Studi di Milano, Dipartimento di Scienze Cliniche e di Comunità, Milan, Italy
| | - Francesco Alamanni
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Università degli Studi di Milano, Dipartimento di Scienze Cliniche e di Comunità, Milan, Italy
| | - Mauro Pepi
- Centro Cardiologico Monzino IRCCS, Milan, Italy
| | | | - Paolo Poggio
- Centro Cardiologico Monzino IRCCS, Milan, Italy.
| |
Collapse
|
22
|
Perrot N, Valerio V, Moschetta D, Boekholdt SM, Dina C, Chen HY, Abner E, Martinsson A, Manikpurage HD, Rigade S, Capoulade R, Mass E, Clavel MA, Le Tourneau T, Messika-Zeitoun D, Wareham NJ, Engert JC, Polvani G, Pibarot P, Esko T, Smith JG, Mathieu P, Thanassoulis G, Schott JJ, Bossé Y, Camera M, Thériault S, Poggio P, Arsenault BJ. Genetic and In Vitro Inhibition of PCSK9 and Calcific Aortic Valve Stenosis. JACC Basic Transl Sci 2020; 5:649-661. [PMID: 32760854 PMCID: PMC7393433 DOI: 10.1016/j.jacbts.2020.05.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/23/2022]
Abstract
The authors investigated whether PCSK9 inhibition could represent a therapeutic strategy in calcific aortic valve stenosis (CAVS). A meta-analysis of 10 studies was performed to determine the impact of the PCSK9 R46L variant on CAVS, and the authors found that CAVS was less prevalent in carriers of this variant (odds ratio: 0.80 [95% confidence interval: 0.70 to 0.91]; p = 0.0011) compared with noncarriers. PCSK9 expression was higher in the aortic valves of patients CAVS compared with control patients. In human valve interstitials cells submitted to a pro-osteogenic medium, PCSK9 levels increased and a PCSK9 neutralizing antibody significantly reduced calcium accumulation.
Collapse
Key Words
- Ad DMEM, advanced Dulbecco’s modified Eagle’s medium
- CAD, coronary artery disease
- CAVS, calcific aortic valve stenosis
- HDL-C, high-density lipoprotein cholesterol
- IQR, interquartile range
- LDL cholesterol
- LDL-C, low-density lipoprotein cholesterol
- Lp(a), lipoprotein(a)
- PBS, phosphate-buffered saline
- PBST, 1× phosphate-buffered saline with 0.1% Triton
- PCSK9, proprotein convertase subtilisin/kexin type 9
- SNP, single nucleotide polymorphism
- TC, total cholesterol
- VIC, valve interstitial cell
- VLDL-C, very-low-density lipoprotein cholesterol
- aortic valve interstitial cell
- apoB, apolipoprotein B
- apolipoprotein B
- calcific aortic valve stenosis
- lipoprotein(a)
- proprotein convertase subtilisin/kexin type 9
- wGRS, weighted genetic risk score
Collapse
Affiliation(s)
- Nicolas Perrot
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Vincenza Valerio
- Centro Cardiologico Monzino IRCCS, Milan, Italy
- Università degli Studi di Napoli Federico II, Dipartimento di Medicina Clinica e Chirurgia, Naples, Italy
| | - Donato Moschetta
- Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Christian Dina
- l'Institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Hao Yu Chen
- McGill University Health Center Research Institute, Montreal, Québec, Canada
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andreas Martinsson
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Hasanga D. Manikpurage
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Sidwell Rigade
- l'Institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Romain Capoulade
- l'Institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Elvira Mass
- University of Bonn, Developmental Biology of the Innate Immune System, Life & Medical Sciences Institute (LIMES), Bonn, Germany
| | - Marie-Annick Clavel
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | | | - David Messika-Zeitoun
- Department of Cardiology, Assistance Publique – Hôpitaux de Paris, Bichat Hospital, Paris, France
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Nicholas J. Wareham
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - James C. Engert
- McGill University Health Center Research Institute, Montreal, Québec, Canada
| | - Gianluca Polvani
- Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Cardiovascular Sciences and Community Health, University of Milan, Milan, Italy
| | - Philippe Pibarot
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - J. Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Patrick Mathieu
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - George Thanassoulis
- McGill University Health Center Research Institute, Montreal, Québec, Canada
| | | | - Yohan Bossé
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Marina Camera
- Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Sébastien Thériault
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | | | - Benoit J. Arsenault
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| |
Collapse
|
23
|
Perrucci GL, Songia P, Moschetta D, Barbagallo VA, Valerio V, Myasoedova VA, Alfieri V, Massaiu I, Roberto M, Malešević M, Pompilio G, Poggio P. Cyclophilin A inhibition as potential treatment of human aortic valve calcification. Pharmacol Res 2020; 158:104888. [PMID: 32434054 DOI: 10.1016/j.phrs.2020.104888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023]
Abstract
Aortic valve stenosis (AS) is a pathological condition that affects about 3% of the population, representing the most common valve disease. The main clinical feature of AS is represented by the impaired leaflet motility, due to calcification, which leads to the left ventricular outflow tract obstruction during systole. The formation and accumulation of calcium nodules are driven by valve interstitial cells (VICs). Unfortunately, to date, the in vitro and in vivo studies were not sufficient to fully recapitulate all the pathological pathways involved in AS development, as well as to define a specific and effective pharmacological treatment for AS patients. Cyclophilin A (CyPA), the most important immunophilin and endogenous ligand of cyclosporine A (CsA), is strongly involved in several detrimental cardiovascular processes, such as calcification. To date, there are no data on the CyPA role in VIC-mediated calcification process of AS. Here, we aimed to identify the role of CyPA in AS by studying VIC calcification, in vitro. In this study, we found that (i) CyPA is up-regulated in stenotic valves of AS patients, (ii) pro-calcifying medium promotes CyPA secretion by VICs, (iii) in vitro treatment of VICs with exogenous CyPA strongly stimulates calcium deposition, and (iv) exogenous CyPA inhibition mediated by CsA analogue MM284 abolished in vitro calcium potential. Thus, CyPA represents a biological target that may act as a novel candidate in the detrimental AS development and its inhibition may provide a novel pharmacological approach for AS treatment.
Collapse
Affiliation(s)
- Gianluca L Perrucci
- Unità di Medicina Rigenerativa e Biologia Vascolare, Centro Cardiologico Monzino IRCCS, Milano, Italy.
| | - Paola Songia
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Donato Moschetta
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milano, Italy; Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Veronica A Barbagallo
- Unità di Medicina Rigenerativa e Biologia Vascolare, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Vincenza Valerio
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milano, Italy; Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Veronika A Myasoedova
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Valentina Alfieri
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Ilaria Massaiu
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Maurizio Roberto
- Dipartimento di Chirurgia Cardiovascolare, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Miroslav Malešević
- Martin-Luther-University Halle-Wittenberg, Institute of Biochemistry and Biotechnology, Enzymology Department, Halle, Germany; Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Giulio Pompilio
- Unità di Medicina Rigenerativa e Biologia Vascolare, Centro Cardiologico Monzino IRCCS, Milano, Italy; Dipartimento di Chirurgia Cardiovascolare, Centro Cardiologico Monzino IRCCS, Milano, Italy; Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Milano, Italy
| | - Paolo Poggio
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milano, Italy.
| |
Collapse
|
24
|
Matilla L, Roncal C, Ibarrola J, Arrieta V, García-Peña A, Fernández-Celis A, Navarro A, Álvarez V, Gainza A, Orbe J, Cachofeiro V, Zalba G, Sádaba R, Rodríguez JA, López-Andrés N. A Role for MMP-10 (Matrix Metalloproteinase-10) in Calcific Aortic Valve Stenosis. Arterioscler Thromb Vasc Biol 2020; 40:1370-1382. [PMID: 32188274 DOI: 10.1161/atvbaha.120.314143] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Aortic valve (AV) calcification plays an important role in the progression of aortic stenosis (AS). MMP-10 (matrix metalloproteinase-10 or stromelysin-2) is involved in vascular calcification in atherosclerosis. We hypothesize that MMP-10 may play a pathophysiological role in calcific AS. Approach and Results: Blood samples (n=112 AS and n=349 controls) and AVs (n=88) from patients undergoing valve replacement were analyzed. Circulating MMP-10 was higher in patients with AS compared with controls (P<0.001) and correlated with TNFα (tumor necrosis factor α; rS=0.451; P<0.0001). MMP-10 was detected by immunochemistry in AVs from patients with AS colocalized with aortic valve interstitial cells markers α-SMA (α-smooth muscle actin) and vimentin and with calcification markers Runx2 (Runt-related transcription factor 2) and SRY (sex-determining region Y)-box 9. MMP-10 expression in AVs was further confirmed by RT-qPCR and western blot. Ex vivo, MMP-10 was elevated in the conditioned media of AVs from patients with AS and associated with interleukin-1β (rS=0.5045, P<0.001) and BMP (bone morphogenetic protein)-2 (rS=0.5003, P<0.01). In vitro, recombinant human MMP-10 induced the overexpression of inflammatory, fibrotic, and osteogenic markers (interleukin-1β, α-SMA, vimentin, collagen, BMP-4, Sox9, OPN [osteopontin], BMP-9, and Smad 1/5/8; P<0.05) and cell mineralization in aortic valve interstitial cells isolated from human AVs, in a mechanism involving Akt (protein kinase B) phosphorylation. These effects were prevented by TIMP-1 (tissue inhibitor of metalloproteinases type 1), a physiological MMP inhibitor, or specifically by an anti-MMP-10 antibody. CONCLUSIONS MMP-10, which is overexpressed in aortic valve from patients with AS, seems to play a central role in calcification in AS through Akt phosphorylation. MMP-10 could be a new therapeutic target for delaying the progression of aortic valve calcification in AS.
Collapse
Affiliation(s)
- Lara Matilla
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - Carmen Roncal
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, IdiSNA, Pamplona, Spain (C.R., J.O., J.A.R.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (C.R., J.O., V.C., J.A.R.)
| | - Jaime Ibarrola
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - Vanessa Arrieta
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - Amaia García-Peña
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - Amaya Fernández-Celis
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - Adela Navarro
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - Virginia Álvarez
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - Alicia Gainza
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - Josune Orbe
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, IdiSNA, Pamplona, Spain (C.R., J.O., J.A.R.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (C.R., J.O., V.C., J.A.R.)
| | - Victoria Cachofeiro
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (C.R., J.O., V.C., J.A.R.).,Departamento de Fisiología, Facultad Medicina, Universidad Complutense, Instituto de Investigacioón Sanitaria Gregorio Maranñoón (IiSGM), Madrid, Spain (V.C.)
| | - Guillermo Zalba
- Department of Biochemistry and Genetics, University of Navarra, IdiSNA, Pamplona, Spain (G.Z.)
| | - Rafael Sádaba
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.)
| | - José A Rodríguez
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, IdiSNA, Pamplona, Spain (C.R., J.O., J.A.R.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (C.R., J.O., V.C., J.A.R.)
| | - Natalia López-Andrés
- From the Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain (L.M., J.I., V. Arrieta, A.G.-P., A.F.-C., A.N., V. Álvarez, A.G., R.S., N.L.-A.).,Université de Lorraine, INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, France (N.L.-A.)
| |
Collapse
|
25
|
Kossar AP, Anselmo W, Grau JB, Liu Y, Small A, Carter SL, Salvador L, Zhao L, Cvijic ME, Li Z, Yarde M, Rioux N, Rader DJ, Levy RJ, Ferrari G. Circulating and tissue matricellular RNA and protein expression in calcific aortic valve disease. Physiol Genomics 2020; 52:191-199. [PMID: 32089075 DOI: 10.1152/physiolgenomics.00104.2019] [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] [Indexed: 11/22/2022] Open
Abstract
Aortic valve sclerosis is a highly prevalent, poorly characterized asymptomatic manifestation of calcific aortic valve disease and may represent a therapeutic target for disease mitigation. Human aortic valve cusps and blood were obtained from 333 patients undergoing cardiac surgery (n = 236 for severe aortic stenosis, n = 35 for asymptomatic aortic valve sclerosis, n = 62 for no valvular disease), and a multiplex assay was used to evaluate protein expression across the spectrum of calcific aortic valve disease. A subset of six valvular tissue samples (n = 3 for asymptomatic aortic valve sclerosis, n = 3 for severe aortic stenosis) was used to create RNA sequencing profiles, which were subsequently organized into clinically relevant gene modules. RNA sequencing identified 182 protein-encoding, differentially expressed genes in aortic valve sclerosis vs. aortic stenosis; 85% and 89% of expressed genes overlapped in aortic stenosis and aortic valve sclerosis, respectively, which decreased to 55% and 84% when we targeted highly expressed genes. Bioinformatic analyses identified six differentially expressed genes encoding key extracellular matrix regulators: TBHS2, SPARC, COL1A2, COL1A1, SPP1, and CTGF. Differential expression of key circulating biomarkers of extracellular matrix reorganization was observed in control vs. aortic valve sclerosis (osteopontin), control vs. aortic stenosis (osteoprotegerin), and aortic valve sclerosis vs. aortic stenosis groups (MMP-2), which corresponded to valvular mRNA expression. We demonstrate distinct mRNA and protein expression underlying aortic valve sclerosis and aortic stenosis. We anticipate that extracellular matrix regulators can serve as circulating biomarkers of early calcific aortic valve disease and as novel targets for early disease mitigation, pending prospective clinical investigations.
Collapse
Affiliation(s)
| | - Wanda Anselmo
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - Juan B Grau
- Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Yichuan Liu
- The Children's Hospital of Philadelphia, Pennsylvania
| | - Aeron Small
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Lei Zhao
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | - Zhuyin Li
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | | | | | - Robert J Levy
- The Children's Hospital of Philadelphia, Pennsylvania
| | - Giovanni Ferrari
- Columbia University, New York, New York.,University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
Heat shock protein 90 is downregulated in calcific aortic valve disease. BMC Cardiovasc Disord 2019; 19:306. [PMID: 31856737 PMCID: PMC6923932 DOI: 10.1186/s12872-019-01294-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 12/03/2019] [Indexed: 01/06/2023] Open
Abstract
Background Calcific aortic valve disease (CAVD) is an atheroinflammatory process; finally it leads to progressive calcification of the valve. There is no effective pharmacological treatment for CAVD and many of the underlying molecular mechanisms remain unknown. We conducted a proteomic study to reveal novel factors associated with CAVD. Methods We compared aortic valves from patients undergoing valvular replacement surgery due to non-calcified aortic insufficiency (control group, n = 5) to a stenotic group (n = 7) using two-dimensional difference gel electrophoresis (2D-DIGE). Protein spots were identified with mass spectrometry. Western blot and immunohistochemistry were used to validate the results in a separate patient cohort and Ingenuity Pathway Analysis (IPA) was exploited to predict the regulatory network of CAVD. Results We detected an upregulation of complement 9 (C9), serum amyloid P-component (APCS) and transgelin as well as downregulation of heat shock protein (HSP90), protein disulfide isomerase A3 (PDIA3), annexin A2 (ANXA2) and galectin-1 in patients with aortic valve stenosis. The decreased protein expression of HSP90 was confirmed with Western blot. Conclusions We describe here a novel data set of proteomic changes associated with CAVD, including downregulation of the pro-inflammatory cytosolic protein, HSP90.
Collapse
|
27
|
Sakaue T, Hamaguchi M, Aono J, Nakashiro KI, Shikata F, Kawakami N, Oshima Y, Kurata M, Nanba D, Masumoto J, Yamaguchi O, Higashiyama S, Izutani H. Valve Interstitial Cell-Specific Cyclooxygenase-1 Associated With Calcification of Aortic Valves. Ann Thorac Surg 2019; 110:40-49. [PMID: 31760051 DOI: 10.1016/j.athoracsur.2019.09.085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 09/10/2019] [Accepted: 09/24/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND The molecular mechanisms underlying aortic valve calcification are poorly understood. Here, we aimed to identify the master regulators of calcification by comparison of genes in valve interstitial cells (VICs) with calcified and noncalcified aortic valves. METHODS Calcified aortic valves were surgically excised from patients with aortic valve stenosis who required aortic valve replacements. Noncalcified and calcified sections were obtained from aortic valve leaflets. Collagenase-digested tissues were seeded into dishes, and VICs adhering to the dishes were cultured for 3 weeks, followed by comprehensive gene expression analysis. Functional analyses of identified proteins were performed by in vitro calcification assays. Tissue localization was determined by immunohistochemical staining for normal (n = 11) and stenotic valves (n = 30). RESULTS We found 87 genes showing greater than a twofold change in calcified tissues. Among these genes, 68 were downregulated and 19 were upregulated. Cyclooxygenase-1 (COX1) messenger RNA and protein levels were upregulated in VICs from calcified tissues. The COX1 messenger RNA and protein levels in VICs were also strongly increased by stimulation with osteoblast differentiation medium. These were VIC-specific phenotypes and were not observed in other cell types. Immunohistochemical staining revealed that COX1-positive VICs were specifically localized in the calcified area of aortic valve tissues. CONCLUSIONS The VIC-specific COX1 overexpression played a crucial role in calcification by promoting osteoblast differentiation in aortic valve tissues.
Collapse
Affiliation(s)
- Tomohisa Sakaue
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Ehime, Japan.
| | - Mika Hamaguchi
- Department of Cardiology, Pulmonology, Hypertension, and Nephrology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun Aono
- Department of Cardiology, Pulmonology, Hypertension, and Nephrology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koh-Ichi Nakashiro
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Ehime University, Ehime, Japan
| | - Fumiaki Shikata
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Ehime, Japan; Queensland Pediatric Cardiac Service, Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Natsuki Kawakami
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yusuke Oshima
- Biomedical Optics Laboratory, Graduate School of Biomedical Engineering Tohoku University, Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi, Japan; Department of Gastroenterological and Pediatric Surgery, Oita University Faculty of Medicine, Idaigaoka, Hasama-machi, Yufu City, Oita, Japan; Oral-Maxillofacial Surgery and Orthodontics, University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Mie Kurata
- Department of Pathology, Division of Analytical Pathology, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Pathology, Proteo-Science Center, Ehime University, Ehime, Japan
| | - Daisuke Nanba
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Junya Masumoto
- Department of Pathology, Division of Analytical Pathology, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Pathology, Proteo-Science Center, Ehime University, Ehime, Japan
| | - Osamu Yamaguchi
- Department of Cardiology, Pulmonology, Hypertension, and Nephrology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Shigeki Higashiyama
- Department of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Ehime, Japan
| | - Hironori Izutani
- Department of Cardiovascular and Thoracic Surgery, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
28
|
Anselmo W, Branchetti E, Grau JB, Li G, Ayoub S, Lai EK, Rioux N, Tovmasyan A, Fortier JH, Sacks MS, Batinic-Haberle I, Hazen SL, Levy RJ, Ferrari G. Porphyrin-Based SOD Mimic MnTnBu OE -2-PyP 5+ Inhibits Mechanisms of Aortic Valve Remodeling in Human and Murine Models of Aortic Valve Sclerosis. J Am Heart Assoc 2019; 7:e007861. [PMID: 30371255 PMCID: PMC6474974 DOI: 10.1161/jaha.117.007861] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Aortic valve sclerosis (AVSc), the early asymptomatic presentation of calcific aortic valve (AV) disease, affects 25% to 30% of patients aged >65 years. In vitro and ex vivo experiments with antioxidant strategies and antagonists of osteogenic differentiation revealed that AVSc is reversible. In this study, we characterized the underlying changes in the extracellular matrix architecture and valve interstitial cell activation in AVSc and tested in vitro and in vivo the activity of a clinically approved SOD (superoxide dismutase) mimic and redox‐active drug MnTnBuOE‐2‐PyP5+ (BMX‐001). Methods and Results After receiving informed consent, samples from patients with AVSc, AV stenosis, and controls were collected. Uniaxial mechanical stimulation and in vitro studies on human valve interstitial cells were performed. An angiotensin II chronic infusion model was used to impose AV thickening and remodeling. We characterized extracellular matrix structures by small‐angle light scattering, scanning electron microscopy, histology, and mass spectrometry. Diseased human valves showed altered collagen fiber alignment and ultrastructural changes in AVSc, accumulation of oxidized cross‐linking products in AV stenosis, and reversible expression of extracellular matrix regulators ex vivo. We demonstrated that MnTnBuOE‐2‐PyP5+ inhibits human valve interstitial cell activation and extracellular matrix remodeling in a murine model (C57BL/6J) of AVSc by electron microscopy and histology. Conclusions AVSc is associated with architectural remodeling despite marginal effects on the mechanical properties in both human and mice. MnTnBuOE‐2‐PyP5+ controls AV thickening in a murine model of AVSc. Because this compound has been approved recently for clinical use, this work could shift the focus for the treatment of calcific AV disease, moving from AV stenosis to an earlier presentation (AVSc) that could be more responsive to medical therapies.
Collapse
Affiliation(s)
| | | | - Juan B Grau
- 2 Ottawa Heart Institute Ottawa Ontario Canada
| | - Gen Li
- 3 Columbia University New York NY
| | | | - Eric K Lai
- 1 University of Pennsylvania Philadelphia PA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Schlotter F, Halu A, Goto S, Blaser MC, Body SC, Lee LH, Higashi H, DeLaughter DM, Hutcheson JD, Vyas P, Pham T, Rogers MA, Sharma A, Seidman CE, Loscalzo J, Seidman JG, Aikawa M, Singh SA, Aikawa E. Spatiotemporal Multi-Omics Mapping Generates a Molecular Atlas of the Aortic Valve and Reveals Networks Driving Disease. Circulation 2019; 138:377-393. [PMID: 29588317 DOI: 10.1161/circulationaha.117.032291] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND No pharmacological therapy exists for calcific aortic valve disease (CAVD), which confers a dismal prognosis without invasive valve replacement. The search for therapeutics and early diagnostics is challenging because CAVD presents in multiple pathological stages. Moreover, it occurs in the context of a complex, multi-layered tissue architecture; a rich and abundant extracellular matrix phenotype; and a unique, highly plastic, and multipotent resident cell population. METHODS A total of 25 human stenotic aortic valves obtained from valve replacement surgeries were analyzed by multiple modalities, including transcriptomics and global unlabeled and label-based tandem-mass-tagged proteomics. Segmentation of valves into disease stage-specific samples was guided by near-infrared molecular imaging, and anatomic layer-specificity was facilitated by laser capture microdissection. Side-specific cell cultures were subjected to multiple calcifying stimuli, and their calcification potential and basal/stimulated proteomes were evaluated. Molecular (protein-protein) interaction networks were built, and their central proteins and disease associations were identified. RESULTS Global transcriptional and protein expression signatures differed between the nondiseased, fibrotic, and calcific stages of CAVD. Anatomic aortic valve microlayers exhibited unique proteome profiles that were maintained throughout disease progression and identified glial fibrillary acidic protein as a specific marker of valvular interstitial cells from the spongiosa layer. CAVD disease progression was marked by an emergence of smooth muscle cell activation, inflammation, and calcification-related pathways. Proteins overrepresented in the disease-prone fibrosa are functionally annotated to fibrosis and calcification pathways, and we found that in vitro, fibrosa-derived valvular interstitial cells demonstrated greater calcification potential than those from the ventricularis. These studies confirmed that the microlayer-specific proteome was preserved in cultured valvular interstitial cells, and that valvular interstitial cells exposed to alkaline phosphatase-dependent and alkaline phosphatase-independent calcifying stimuli had distinct proteome profiles, both of which overlapped with that of the whole tissue. Analysis of protein-protein interaction networks found a significant closeness to multiple inflammatory and fibrotic diseases. CONCLUSIONS A spatially and temporally resolved multi-omics, and network and systems biology strategy identifies the first molecular regulatory networks in CAVD, a cardiac condition without a pharmacological cure, and describes a novel means of systematic disease ontology that is broadly applicable to comprehensive omics studies of cardiovascular diseases.
Collapse
Affiliation(s)
- Florian Schlotter
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Arda Halu
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.).,Channing Division of Network Medicine (A.H., A.S., M.A.)
| | - Shinji Goto
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Simon C Body
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA. Center for Perioperative Genomics and Department of Anesthesiology, Brigham and Women's Hospital, Boston, MA (S.C.B.)
| | - Lang H Lee
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Daniel M DeLaughter
- Department of Genetics, Harvard Medical School, Boston, MA (D.M.D., C.E.S., J.G.S.)
| | - Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.).,Department of Biomedical Engineering, Florida International University, Miami (J.D.H.)
| | - Payal Vyas
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Maximillian A Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Amitabh Sharma
- Channing Division of Network Medicine (A.H., A.S., M.A.)
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (D.M.D., C.E.S., J.G.S.).,Department of Medicine, Brigham and Women's Hospital, Boston, MA (C.E.S., J.L.).,Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Boston, MA (C.E.S., J.L.)
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA (D.M.D., C.E.S., J.G.S.)
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.).,Channing Division of Network Medicine (A.H., A.S., M.A.).,Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.A., E.A.)
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.)
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine (F.S., A.H., S.G., M.C.B., L.H.L., H.H., J.D.H., P.V., T.P., M.A.R., M.A., S.A.S., E.A.).,Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.A., E.A.)
| |
Collapse
|
30
|
Aortic Valve Sclerosis Adds to Prediction of Short-Term Mortality in Patients with Documented Coronary Atherosclerosis. J Clin Med 2019; 8:jcm8081172. [PMID: 31387275 PMCID: PMC6722808 DOI: 10.3390/jcm8081172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/15/2019] [Accepted: 07/29/2019] [Indexed: 11/25/2022] Open
Abstract
Aims: Aortic valve sclerosis (AVSc), a non-uniform thickening of leaflets with an unrestricted opening, is characterized by inflammation, lipoprotein deposition, and matrix degradation. In the general population, AVSc predicts long-term cardiovascular mortality (+50%) even after adjustment for vascular risk factors and clinical atherosclerosis. We have hypothesized that AVSc is a risk-multiplier able to predict even short-term mortality. To address this issue, we retrospectively analyzed 90-day mortality of all patients who underwent isolated coronary artery bypass grafting (CABG) at Centro Cardiologico Monzino over a ten-year period (2006–2016). Methods: We analyzed 2246 patients and 90-day all-cause mortality was 1.5% (31 deaths). We selected only patients deceased from cardiac causes (n = 29) and compared to alive patients (n = 2215). A cardiologist classified the aortic valve as no-AVSc (n = 1352) or AVSc (n = 892). Cox linear regression and integrated discrimination improvement (IDI) analyses were used to evaluate AVSc in predicting 90-day mortality. Results: AVSc 90-day survival (97.6%) was lower than in no-AVSc (99.4%; p < 0.0001) with a hazard ratio (HR) of 4.0 (95%CI: 1.78, 9.05; p < 0.0001). The HR for AVSc, adjusted for propensity score, was 2.7 (95%CI: 1.17, 6.23; p = 0.02) and IDI statistics confirmed that AVSc significantly adds (p < 0.001) to the identification of high-risk patients than EuroSCORE II alone. Conclusion: Our data supports the hypothesis that a risk stratification strategy based on AVSc, added to ESII, may allow better recognition of patients at high-risk of short-term mortality after isolated surgical myocardial revascularization. Results from this study warrant further confirmation.
Collapse
|
31
|
Baugh L, Watson MC, Kemmerling EC, Hinds PW, Huggins GS, Black LD. Knockdown of CD44 expression decreases valve interstitial cell calcification in vitro. Am J Physiol Heart Circ Physiol 2019; 317:H26-H36. [PMID: 30951363 PMCID: PMC6692733 DOI: 10.1152/ajpheart.00123.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/23/2022]
Abstract
The lack of pharmaceutical targets available to treat patients with calcific aortic valve disease (CAVD) necessitates further research into the specific mechanisms of the disease. The significant changes that occur to the aortic valves extracellular matrix (ECM) during the progression of CAVD suggests that these proteins may play an important role in calcification. Exploring the relationship between valve interstitial cells (VICs) and the ECM may lead to a better understand of CAVD mechanisms and potential pharmaceutical targets. In this study, we look at the effect of two ECM components, collagen and hyaluronic acid (HA), on the mineralization of VICs within the context of a two-dimensional, polyacrylamide (PAAM) model system. Using a novel, nondestructive imaging technique, we were able to track calcific nodule development in culture systems over a 3-wk time frame. We saw a significant increase in the size of the nodules grown on HA PAAM gels as compared with collagen PAAM gels, suggesting that HA has a direct effect on mineralization. Directly looking at the two known receptors of HA, CD44 and receptor for HA-mediated motility (RHAMM), and using siRNA knockdown revealed that a decrease in CD44 expression resulted in a reduction of calcification. A decrease in CD44, through siRNA knockdown, reduces mineralization on HA PAAM gels, suggesting a potential new target for CAVD treatment. NEW & NOTEWORTHY Our in vitro model of calcific aortic valve disease shows an interaction between the hyaluronic acid binding protein CD44 with the osteogenic factor OPN as a potential mechanism of aortic valve calcification. Using siRNA knockdown of CD44, we show an upregulation of OPN expression with a decrease in overall mineralization.
Collapse
Affiliation(s)
- Lauren Baugh
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
| | - Matthew C Watson
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
- Department of Mechanical Engineering, Tufts University , Medford, Massachusetts
| | - Erica C Kemmerling
- Department of Mechanical Engineering, Tufts University , Medford, Massachusetts
| | - Philip W Hinds
- Cellular, Molecular, and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine , Boston, Massachusetts
| | - Gordon S Huggins
- Molecular Cardiology Research Center, Tufts Medical Center and Tufts University Sackler School for Graduate Biomedical Sciences , Boston, Massachusetts
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine , Boston, Massachusetts
| |
Collapse
|
32
|
Poggio P, Songia P, Moschetta D, Valerio V, Myasoedova V, Perrucci GL, Pompilio G. MiRNA profiling revealed enhanced susceptibility to oxidative stress of endothelial cells from bicuspid aortic valve. J Mol Cell Cardiol 2019; 131:146-154. [PMID: 31026425 DOI: 10.1016/j.yjmcc.2019.04.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/15/2019] [Accepted: 04/22/2019] [Indexed: 12/15/2022]
Abstract
AIMS Calcific aortic valve stenosis (CAVS) is the most frequent manifestation of aortic valve disease and the third leading cause of cardiovascular disease in the Western countries associated with significant morbidity and mortality. An active biological progression involving inflammation and oxidation leading to valve endothelial damage is considered a hallmark of the early stages of valve degeneration. However, tricuspid (TAV) and bicuspid (BAV) aortic valve deterioration are considered to differ only by shear stress. We hypothesized that endothelial cells (EC) derived from BAV and TAV patients have different miRNA expression patterns and thus distinct pathways could lead to endothelial damage in BAV than TAV patients. METHODS AND RESULTS We isolated ECs from patients with bicuspid or tricuspid aortic valve, which underwent surgery due to CAVS. MiRNA expression profile by PCR revealed eight upregulated miRNAs between BAV and TAV ECs. Functional analysis identified that BAV ECs presented altered cellular response to oxidative stress and DNA damage stimulus via p53 and alteration in the intrinsic apoptotic signaling pathway. GPX3 and SRXN1 mRNA were express at lower levels in BAV compared to TAV ECs, leading to an increment of DNA double-strand breaks. BAV ECs had a sustained apoptosis activation when compared to TAV ECs. This difference was exacerbated by oxidative stress stimulus leading to a reduced survival rate but completely reverted by miR-328-3p inhibition. CONCLUSION The present data showed molecular differences in oxidative stress susceptibility, DNA damage magnitude, and apoptosis induction between ECs derived from BAV and TAV patients.
Collapse
Affiliation(s)
- Paolo Poggio
- Centro Cardiologico Monzino IRCCS, Unità per lo Studio di Patologie Aortiche, Valvolari e Coronariche, Milan, Italy.
| | - Paola Songia
- Centro Cardiologico Monzino IRCCS, Unità per lo Studio di Patologie Aortiche, Valvolari e Coronariche, Milan, Italy
| | - Donato Moschetta
- Centro Cardiologico Monzino IRCCS, Unità per lo Studio di Patologie Aortiche, Valvolari e Coronariche, Milan, Italy
| | - Vincenza Valerio
- Centro Cardiologico Monzino IRCCS, Unità per lo Studio di Patologie Aortiche, Valvolari e Coronariche, Milan, Italy; Università degli Studi di Napoli Federico II, Dipartimento di Medicina Clinica e Chirurgia, Napoli, Italy
| | - Veronika Myasoedova
- Centro Cardiologico Monzino IRCCS, Unità per lo Studio di Patologie Aortiche, Valvolari e Coronariche, Milan, Italy
| | - Gianluca L Perrucci
- Centro Cardiologico Monzino IRCCS, Unità di Medicina Rigenerativa e Biologia Vascolare, Milan, Italy
| | - Giulio Pompilio
- Centro Cardiologico Monzino IRCCS, Unità di Medicina Rigenerativa e Biologia Vascolare, Milan, Italy; Università degli Studi di Milano, Dipartimento di Scienze Cliniche e di Comunità, Milan, Italy; Centro Cardioloigco Monzino IRCCS, Dipartimento di Chirurgia Cardiovascolare, Milan, Italy.
| |
Collapse
|
33
|
Di Minno MND, Di Minno A, Ambrosino P, Songia P, Pepi M, Tremoli E, Poggio P. Cardiovascular morbidity and mortality in patients with aortic valve sclerosis: A systematic review and meta-analysis. Int J Cardiol 2018; 260:138-144. [PMID: 29622430 DOI: 10.1016/j.ijcard.2018.01.054] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/18/2017] [Accepted: 01/15/2018] [Indexed: 11/17/2022]
Abstract
AIMS The association between aortic valve sclerosis (AVSc) and cardiovascular (CV) events is not consistent among different studies. We have performed a meta-analysis evaluating the association between AVSc and fatal and/or non-fatal CV and cerebrovascular events. METHODS AND RESULTS A systematic search was performed in the electronic databases (PubMed, Web of Science, Scopus, EMBASE). Studies evaluating coronary artery disease (CAD), stroke and CV mortality in AVSc patients and controls were included. Differences among cases and controls were expressed as Odds Ratio (OR) with pertinent 95% Confidence Intervals (CI). Thirty-one studies on 10,537 AVSc patients and 25,005 controls were included in the final analysis. The absolute risk of CAD was 45.8% (95% CI: 32.9-59.3) in AVSc patients and 29.4% (95% CI: 21.8-38.5) in controls with an OR of 2.02 (95% CI: 1.67-2.44) and an attributable risk of 35.8%. Moreover, stroke was reported in 11.8% (95% CI: 4.4-27.7) of AVSc patients and 7.9% (95% CI: 2.5-22.7) of controls (OR: 1.41, 95% CI: 1.16-1.71) with an attributable risk of 33.0%. CV mortality was 6.2% (95% CI: 2.7-13.5) in AVSc patients and 2.0% (95% CI: 0.5-7.9) in controls (OR: 2.70, 95% CI: 1.45-5.01), with an attributable risk of 67.7%. Results were confirmed when pooling together ORs for CAD, stroke and CV mortality obtained by means of multivariate analysis. CONCLUSIONS AVSc is associated with CAD, stroke and CV mortality. Taken together, these data suggest that patients with AVSc may benefit from a stricter CV risk monitoring and that AVSc screening may be included in the frame of CV risk stratification protocols.
Collapse
Affiliation(s)
| | - Alessandro Di Minno
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; Dipartimento di Scienze Farmacologiche e Biomolecolare, University of Milan, Milan, Italy
| | - Pasquale Ambrosino
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Paola Songia
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Mauro Pepi
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | - Paolo Poggio
- Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| |
Collapse
|
34
|
Mazur P, Mielimonka A, Natorska J, Wypasek E, Gawęda B, Sobczyk D, Kapusta P, Malinowski KP, Kapelak B. Lymphocyte and monocyte subpopulations in severe aortic stenosis at the time of surgical intervention. Cardiovasc Pathol 2018; 35:1-7. [PMID: 29727769 DOI: 10.1016/j.carpath.2018.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Aortic stenosis (AS) is the most common acquired valvular heart disease in adults. Immune system involvement becomes evident during AS development. We sought to investigate the role of different circulating lymphocyte and monocyte subpopulations, with focus on CD4+CD8+ and natural killer T (NKT) cells, in AS. MATERIAL AND METHODS Blood samples and aortic valves were obtained from patients undergoing elective aortic valve surgery. Valves were dissected and underwent genetic analyses and calcium content assessment. Lymphocytes and monocytes subsets were assessed by flow cytometry. RESULTS Thirty-eight AS patients were studied. Maximal transvalvular pressure gradient (PGmax) as well as mean transvalvular pressure gradient (PGmean) correlated with the CD4+CD8+ lymphocyte count (r=0.35, P=.03 and r=0.43, P=.006, respectively) and fraction (r=0.43, P=.007 and r=0.48, P=.002, respectively). PGmax and PGmean correlated with CD16+CD56+CD3+ NKT cell count (r=0.39, P=.01 and r=0.43, P=.007, respectively) and fraction (r=0.49, P=.002 and r=0.47, P=.003, respectively). The classical monocyte subpopulation increased after the surgery by 68% (P<.0001). Patients after mini-sternotomy surgery had 47% lower nonclassical monocyte counts than those with full-sternotomy (P=.03). Patients treated with statins had significantly lower postoperative levels of both classical (-25%, P=.04) and nonclassical monocytes (-37%, P=.004) than nontreated individuals. CONCLUSIONS In patients with severe isolated AS, CD4+CD8+ T cells and CD16+CD56+CD3+ NKT cells are associated with AV pressure gradients. Postoperative monocyte levels are affected by procedure invasiveness and use of statins.
Collapse
Affiliation(s)
- Piotr Mazur
- Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland; The John Paul II Hospital, Krakow, Poland.
| | | | - Joanna Natorska
- Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland; The John Paul II Hospital, Krakow, Poland
| | - Ewa Wypasek
- Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland; The John Paul II Hospital, Krakow, Poland; Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, Krakow, Poland
| | | | | | | | | | - Bogusław Kapelak
- Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland; The John Paul II Hospital, Krakow, Poland
| |
Collapse
|
35
|
Driesbaugh KH, Branchetti E, Grau JB, Keeney SJ, Glass K, Oyama MA, Rioux N, Ayoub S, Sacks MS, Quackenbush J, Levy RJ, Ferrari G. Serotonin receptor 2B signaling with interstitial cell activation and leaflet remodeling in degenerative mitral regurgitation. J Mol Cell Cardiol 2017; 115:94-103. [PMID: 29291394 DOI: 10.1016/j.yjmcc.2017.12.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 11/17/2022]
Abstract
AIMS Mitral valve interstitial cells (MVIC) play an important role in the pathogenesis of degenerative mitral regurgitation (MR) due to mitral valve prolapse (MVP). Numerous clinical studies have observed serotonin (5HT) dysregulation in cardiac valvulopathies; however, the impact of 5HT-mediated signaling on MVIC activation and leaflet remodeling in MVP have been investigated to a limited extent. Here we test the hypothesis that 5HT receptors (5HTRs) signaling contributes to MVP pathophysiology. METHODS AND RESULTS Diseased human MV leaflets were obtained during cardiac surgery for MVP; normal MV leaflets were obtained from heart transplants. MV RNA was used for microarray analysis of MVP patients versus control, highlighting genes that indicate the involvement of 5HTR pathways and extracellular matrix remodeling in MVP. Human MV leaflets were also studied in vitro and ex vivo with biomechanical testing to assess remodeling in the presence of a 5HTR2B antagonist (LY272015). MVP leaflets from Cavalier King Charles Spaniels were used as a naturally acquired in vivo model of MVP. These canine MVP leaflets (N=5/group) showed 5HTR2B upregulation. This study also utilized CB57.1ML/6 mice in order to determine the effect of Angiotensin II infusion on MV remodeling. Histological analysis showed that MV thickening due to chronic Angiotensin II remodeling is mitigated by a 5HTR2B antagonist (LY272015) but not by 5HTR2A inhibitors. CONCLUSION In humans, MVP is associated with an upregulation in 5HTR2B expression and increased 5HT receptor signaling in the leaflets. Antagonism of 5HTR2B mitigates MVIC activation in vitro and MV remodeling in vivo. These observations support the view that 5HTR signaling is involved not only in previously reported 5HT-related valvulopathies, but it is also involved in the pathological remodeling of MVP.
Collapse
Affiliation(s)
| | | | - Juan B Grau
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Samuel J Keeney
- University of Pennsylvania, Philadelphia, PA, United States; Columbia University, New York, NY, United States
| | - Kimberly Glass
- Dana Farber Cancer Center, Harvard University, Boston, MT, United States
| | - Mark A Oyama
- University of Pennsylvania, Philadelphia, PA, United States
| | - Nancy Rioux
- The Valley Hospital, Ridgewood, NJ, United States
| | - Salma Ayoub
- University of Texas at Austin, Austin, TX, United States
| | | | - John Quackenbush
- Dana Farber Cancer Center, Harvard University, Boston, MT, United States
| | - Robert J Levy
- The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | | |
Collapse
|
36
|
Munjal C, Jegga AG, Opoka AM, Stoilov I, Norris RA, Thomas CJ, Smith JM, Mecham RP, Bressan GM, Hinton RB. Inhibition of MAPK-Erk pathway in vivo attenuates aortic valve disease processes in Emilin1-deficient mouse model. Physiol Rep 2017; 5:5/5/e13152. [PMID: 28270590 PMCID: PMC5350168 DOI: 10.14814/phy2.13152] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/08/2017] [Indexed: 12/20/2022] Open
Abstract
Aortic valve disease (AVD) is a common condition with a progressive natural history, and presently, there are no pharmacologic treatment strategies. Elastic fiber fragmentation (EFF) is a hallmark of AVD, and increasing evidence implicates developmental elastic fiber assembly defects. Emilin1 is a glycoprotein necessary for elastic fiber assembly that is present in both developing and mature human and mouse aortic valves. The Emilin1‐deficient mouse (Emilin1−/−) is a model of latent AVD, characterized by activated TGFβ/MEK/p‐Erk signaling and upregulated elastase activity. Emilin1−/− aortic valves demonstrate early EFF and aberrant angiogenesis followed by late neovascularization and fibrosis. The objective of this study was to test the effectiveness of three different targeted therapies. Aged (12–14 months) Emilin1−/− mice were treated with refametinib (RDEA‐119, MEK1/2 inhibitor), doxycycline (elastase inhibitor), or G6‐31 (anti‐VEGF‐A mouse antibody) for 4 weeks. Refametinib‐ and doxycycline‐treated Emilin1−/− mice markedly reduced MEK/p‐Erk activation in valve tissue. Furthermore, both refametinib and doxycycline attenuated elastolytic cathepsin K, L, MMP‐2, and MMP‐9 activation, and abrogated macrophage and neutrophil infiltration in Emilin1−/− aortic valves. RNAseq analysis was performed in aortic valve tissue from adult (4 months) and aged (14 months) Emilin1−/− and age‐matched wild‐type control mice, and demonstrated upregulation of genes associated with MAPK/MEK/p‐Erk signaling and elastases at the adult stage and inflammatory pathways at the aged stage controlling for age. These results suggest that Erk1/2 signaling is an important modulator of early elastase activation, and pharmacological inhibition using refametinib may be a promising treatment to halt AVD progression
Collapse
Affiliation(s)
- Charu Munjal
- Divisions of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Anil G Jegga
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Amy M Opoka
- Divisions of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ivan Stoilov
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Ohio
| | - Russell A Norris
- Department of Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Craig J Thomas
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences National Institutes of Health, Bethesda, Maryland
| | - J Michael Smith
- TriHealth Heart Institute, Cardio-Thoracic Surgery, Cincinnati, Ohio
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Ohio
| | - Giorgio M Bressan
- The Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Robert B Hinton
- Divisions of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
37
|
Different Notch signaling in cells from calcified bicuspid and tricuspid aortic valves. J Mol Cell Cardiol 2017; 114:211-219. [PMID: 29158034 DOI: 10.1016/j.yjmcc.2017.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/13/2017] [Indexed: 12/11/2022]
Abstract
AIMS Calcific aortic valve disease is the most common heart valve disease in the Western world. Bicuspid and tricuspid aortic valve calcifications are traditionally considered together although the dynamics of the disease progression is different between the two groups of patients. Notch signaling is critical for bicuspid valve development and NOTCH1 mutations are associated with bicuspid valve and calcification. We hypothesized that Notch-dependent mechanisms of valve mineralization might be different in the two groups. METHODS AND RESULTS We used aortic valve interstitial cells and valve endothelial cells from patients with calcific aortic stenosis with bicuspid or tricuspid aortic valve. Expression of Notch-related genes in valve interstitial cells by qPCR was different between bicuspid and tricuspid groups. Discriminant analysis of gene expression pattern in the interstitial cells revealed that the cells from calcified bicuspid valves formed a separate group from calcified tricuspid and control cells. Interstitial cells from bicuspid calcified valves demonstrated significantly higher sensitivity to stimuli at early stages of induced proosteogenic differentiation and were significantly more sensitive to the activation of proosteogenic OPN, ALP and POSTIN expression by Notch activation. Notch-activated endothelial-to-mesenchymal transition and the corresponding expression of HEY1 and SLUG were also more prominent in bicuspid valve derived endothelial cells compared to the cells from calcified tricuspid and healthy valves. CONCLUSION Early signaling events including Notch-dependent mechanisms that are responsible for the initiation of aortic valve calcification are different between the patients with bicuspid and tricuspid aortic valves.
Collapse
|
38
|
Krishnamurthy VK, Stout AJ, Sapp MC, Matuska B, Lauer ME, Grande-Allen KJ. Dysregulation of hyaluronan homeostasis during aortic valve disease. Matrix Biol 2017; 62:40-57. [PMID: 27856308 PMCID: PMC10615645 DOI: 10.1016/j.matbio.2016.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 11/05/2016] [Accepted: 11/08/2016] [Indexed: 01/03/2023]
Abstract
Aortic valve disease (AVD) is one of the leading causes of cardiovascular mortality. Abnormal expression of hyaluronan (HA) and its synthesizing/degrading enzymes have been observed during latent AVD however, the mechanism of impaired HA homeostasis prior to and after the onset of AVD remains unexplored. Transforming growth factor beta (TGFβ) pathway defects and biomechanical dysfunction are hallmarks of AVD, however their association with altered HA regulation is understudied. Expression of HA homeostatic markers was evaluated in diseased human aortic valves and TGFβ1-cultured porcine aortic valve tissues using histology, immunohistochemistry and Western blotting. Further, porcine valve interstitial cell cultures were stretched (using Flexcell) and simultaneously treated with exogenous TGFβ1±inhibitors for activated Smad2/3 (SB431542) and ERK1/2 (U0126) pathways, and differential HA regulation was assessed using qRT-PCR. Pathological heavy chain HA together with abnormal regional expression of the enzymes HAS2, HYAL1, KIAA1199, TSG6 and IαI was demonstrated in calcified valve tissues identifying the collapse of HA homeostatic machinery during human AVD. Heightened TSG6 activity likely preceded the end-stage of disease, with the existence of a transitional, pre-calcific phase characterized by HA dysregulation. TGFβ1 elicited a fibrotic remodeling response in porcine aortic valves similar to human disease pathology, with increased collagen and HYAL to HAS ratio, and site-specific abnormalities in the expression of CD44 and RHAMM receptors. Further in these porcine valves, expression of HAS2 and HYAL1 was found to be differentially regulated by the Smad2/3 and ERK1/2 pathways, and CD44 expression was highly responsive to biomechanical strain. Leveraging the regulatory pathways that control both HA maintenance in normal valves and early postnatal dysregulation of HA homeostasis during disease may identify new mechanistic insight into AVD pathogenesis.
Collapse
Affiliation(s)
| | - Andrew J Stout
- Department of Materials Science and Nanoengineering, Rice University, Houston, TX 77005, USA
| | - Matthew C Sapp
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Brittany Matuska
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mark E Lauer
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
39
|
Perrucci GL, Zanobini M, Gripari P, Songia P, Alshaikh B, Tremoli E, Poggio P. Pathophysiology of Aortic Stenosis and Mitral Regurgitation. Compr Physiol 2017. [PMID: 28640443 DOI: 10.1002/cphy.c160020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The global impact of the spectrum of valve diseases is a crucial, fast-growing, and underrecognized health problem. The most prevalent valve diseases, requiring surgical intervention, are represented by calcific and degenerative processes occurring in heart valves, in particular, aortic and mitral valve. Due to the increasing elderly population, these pathologies will gain weight in the global health burden. The two most common valve diseases are aortic valve stenosis (AVS) and mitral valve regurgitation (MR). AVS is the most commonly encountered valve disease nowadays and affects almost 5% of elderly population. In particular, AVS poses a great challenge due to the multiple comorbidities and frailty of this patient subset. MR is also a common valve pathology and has an estimated prevalence of 3% in the general population, affecting more than 176 million people worldwide. This review will focus on pathophysiological changes in both these valve diseases, starting from the description of the anatomical aspects of normal valve, highlighting all the main cellular and molecular features involved in the pathological progression and cardiac consequences. This review also evaluates the main approaches in clinical management of these valve diseases, taking into account of the main published clinical guidelines. © 2017 American Physiological Society. Compr Physiol 7:799-818, 2017.
Collapse
Affiliation(s)
- Gianluca L Perrucci
- Centro Cardiologico Monzino, IRCCS, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | | | - Paola Songia
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | | - Paolo Poggio
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| |
Collapse
|
40
|
Human interstitial cellular model in therapeutics of heart valve calcification. Amino Acids 2017; 49:1981-1997. [DOI: 10.1007/s00726-017-2432-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/27/2017] [Indexed: 12/27/2022]
|
41
|
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: 56] [Impact Index Per Article: 7.0] [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
|
42
|
Songia P, Branchetti E, Parolari A, Myasoedova V, Ferrari G, Alamanni F, Tremoli E, Poggio P. Mitral valve endothelial cells secrete osteoprotegerin during endothelial mesenchymal transition. J Mol Cell Cardiol 2016; 98:48-57. [PMID: 27338002 DOI: 10.1016/j.yjmcc.2016.06.061] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 06/16/2016] [Accepted: 06/18/2016] [Indexed: 12/20/2022]
Abstract
AIMS Mitral valve prolapse (MVP) has a prevalence of 3% in the general population, affecting >176 million people worldwide. Despite this, little is known about the molecular and cellular mechanisms involved in the progression of MVP and surgical intervention is the only available option. In this study we investigated the role of osteoprotegerin (OPG) during endothelial to mesenchymal transition (EndMT) in MVP. METHODS AND RESULTS VECs and VICs were isolated from posterior mitral valve leaflets of patients undergoing mitral valve repair (n=25). Plasma was collected from 57 subjects (29 controls and 28 MVP patients). Overexpression of OPG during EndMT followed by autocrine effects characterised by reactive oxygen species increment and accelerated migration was documented. In addition, OPG increased VIC proliferation. Finally, OPG plasma levels were significantly higher in MVP patients compared to control subjects and the area under the ROC curve was 0.92. CONCLUSION EndMT has been recognised as a possible pathological mechanism for MVP. For the first time, we report the involvement of OPG in cellular and molecular changes in MVP isolated cells. In addition, we detected elevated circulating OPG levels in MVP patients when compared to controls, which supports the hypothesis that OPG is involved in MVP development and progression.
Collapse
Affiliation(s)
- Paola Songia
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Università degli Studi di Milano, Dipartimento di Scienze Farmacologiche e Biomolecolari, Milan, Italy
| | | | - Alessandro Parolari
- Policlinico San Donato IRCCS, U.O. Cardiochirurgia e Ricerca traslazionale, San Donato Milanese, Italy; Università degli Studi di Milano, Dipartimento di Scienze Biomediche per la Salute, Milan, Italy
| | | | - Giovanni Ferrari
- University of Pennsylvania, Department of Surgery, Philadelphia, PA, USA
| | - Francesco Alamanni
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Università degli Studi di Milano, Dipartimento di Scienze Cliniche e di Comunità, Sezione Cardiovascolare, Milan, Italy
| | | | - Paolo Poggio
- Centro Cardiologico Monzino IRCCS, Milan, Italy.
| |
Collapse
|
43
|
Di Minno MND, Di Minno A, Ambrosino P, Songia P, Tremoli E, Poggio P. Aortic valve sclerosis as a marker of atherosclerosis: Novel insights from hepatic steatosis. Int J Cardiol 2016; 217:1-6. [PMID: 27164417 DOI: 10.1016/j.ijcard.2016.04.162] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 04/29/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Nowadays, aortic valve sclerosis (AVSc) might be considered an atherosclerosis-like process due to significant association with age, male gender and some major features of metabolic syndrome. Nonalcoholic fatty liver disease (NAFLD) has been recognized as a clinical expression of the metabolic syndrome and as a predictor of cardiovascular events. We aim, with this meta-analysis, to evaluate the correlation between NAFLD and AVSc; this finding might suggest new insights and interactions among NAFLD, AVSc and the atherosclerotic process. METHODS AND RESULTS A detailed search was conducted according to Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines to identify all studies providing data about the association between AVSc and NAFLD. 3 studies enrolling a total of 1172 patients with NAFLD and 1467 controls without NAFLD were included in the meta-analysis. The prevalence of AVSc was 41.3% (95% CI: 32.0%, 51.4%) in NAFLD patients and 24.6% (18.4%, 32.0%) in subjects without NAFLD with a corresponding OR of 2.28 (95% CI: 1.21, 4.28, p=0.01, I(2): 77.6%, p=0.011). A meta-regression analysis showed that age, prevalence of male gender, hypertension, body mass index, and dyslipidemia were directly and significantly associated with the difference in the prevalence of AVSc between patients with NAFLD and those without. CONCLUSIONS In conclusion, our meta-analysis shows a significant association between NAFLD and AVSc. However, further evidence is needed to validate these findings and find out if there is a real link or just a mere association.
Collapse
Affiliation(s)
- Matteo Nicola Dario Di Minno
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; Department of Advanced Biomedical Sciences, Division of Cardiology, Federico II University, Naples, Italy
| | - Alessandro Di Minno
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; University of Milan, Department of Pharmacological and Biomolecular Sciences, Milan, Italy
| | - Pasquale Ambrosino
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Paola Songia
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; University of Milan, Department of Pharmacological and Biomolecular Sciences, Milan, Italy
| | | | - Paolo Poggio
- Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| |
Collapse
|
44
|
Latif N, Quillon A, Sarathchandra P, McCormack A, Lozanoski A, Yacoub MH, Chester AH. Modulation of human valve interstitial cell phenotype and function using a fibroblast growth factor 2 formulation. PLoS One 2015; 10:e0127844. [PMID: 26042674 PMCID: PMC4456368 DOI: 10.1371/journal.pone.0127844] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/21/2015] [Indexed: 12/04/2022] Open
Abstract
Valve interstitial cells (VICs) are fibroblastic in nature however in culture it is widely accepted that they differentiate into a myofibroblastic phenotype. This study assessed a fibroblast culture media formulation for its ability to maintain the phenotype and function of VICs as in the intact healthy valve. Normal human VICs were cultured separately in standard DMEM and in fibroblast media consisting of FGF2 (10ng/ml), insulin (50ng/ml) and 2% FCS for at least a week. Cell morphology, aspect ratio, size, levels and distribution of protein expression, proliferation, cell cycle, contraction and migration were assessed. Some VICs and some valve endothelial cells expressed FGF2 in valve tissue and this expression was increased in calcified valves. VICs in DMEM exhibited large, spread cells whereas VICs in fibroblast media were smaller, elongated and spindly. Aspect ratio and size were both significantly higher in DMEM (p<0.01). The level of expression of α-SMA was significantly reduced in fibroblast media at day 2 after isolation (p<0.01) and the expression of α-SMA, SM22 and EDA-fibronectin was significantly reduced in fibroblast media at days 7 and 12 post-isolation (p<0.01). Expression of cytoskeletal proteins, bone marker proteins and extracellular matrix proteins was reduced in fibroblast media. Proliferation of VICs in fibroblast media was significantly reduced at weeks 1 (p<0.05) and 2 (p<0.01). Collagen gel contraction was significantly reduced in fibroblast media (p<0.05). VICs were found to have significantly fewer and smaller focal adhesions in fibroblast media (p<0.01) with significantly fewer supermature focal adhesions in fibroblast media (p<0.001). Ultrastructurally, VICs in fibroblast media resembled native VICs from intact valves. VICs in fibroblast media demonstrated a slower migratory ability after wounding at 72 hours (p<0.01). Treatment of human VICs with this fibroblast media formulation has the ability to maintain and to dedifferentiate the VICs back to a fibroblastic phenotype with phenotypic and functional characteristics ascribed to cells in the intact valve. This methodology is fundamental in the study of normal valve biology, pathology and in the field of tissue engineering.
Collapse
Affiliation(s)
- Najma Latif
- Imperial College, Heart Science Centre, Harefield Hospital, Harefield, Middx, UB9 6JH, United Kingdom
- QCRC, Qatar Cardiovascular Research Centre, Qatar Foundation, Doha, Qatar
- * E-mail:
| | - Alfred Quillon
- Département de Biologie, École Normale Supérieure de Lyon, Université de Lyon, UCB Lyon1, 46 Allée d’Italie, Lyon, France
| | - Padmini Sarathchandra
- Imperial College, Heart Science Centre, Harefield Hospital, Harefield, Middx, UB9 6JH, United Kingdom
| | - Ann McCormack
- Imperial College, Heart Science Centre, Harefield Hospital, Harefield, Middx, UB9 6JH, United Kingdom
| | - Alec Lozanoski
- Imperial College, Heart Science Centre, Harefield Hospital, Harefield, Middx, UB9 6JH, United Kingdom
| | - Magdi H. Yacoub
- Imperial College, Heart Science Centre, Harefield Hospital, Harefield, Middx, UB9 6JH, United Kingdom
- QCRC, Qatar Cardiovascular Research Centre, Qatar Foundation, Doha, Qatar
| | - Adrian H. Chester
- Imperial College, Heart Science Centre, Harefield Hospital, Harefield, Middx, UB9 6JH, United Kingdom
- QCRC, Qatar Cardiovascular Research Centre, Qatar Foundation, Doha, Qatar
| |
Collapse
|