1
|
Men S, Yu Z, Liu X, Daitoku K, Tachizaki M, Kawaguchi S, Imaizumi T, Minakawa M, Seya K. Role of CD34 in calcification of human aortic valve interstitial cells from patients with aortic valve stenosis. J Pharmacol Sci 2024; 156:198-207. [PMID: 39313278 DOI: 10.1016/j.jphs.2024.09.002] [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: 04/30/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Various osteogenic factors are involved in ectopic human aortic valve calcification; however, the key cell species involved in calcification remains unclear. In a previous study, we reported that mesenchymal stem (CD73, 90, 105) and endothelial (VEGFR2) cell markers are positive in almost all human aortic valve interstitial cells (HAVICs) obtained from a patient with calcified aortic valve stenosis (CAVS). Further, CD34-negative HAVICs are highly sensitive to calcification stimulations. Here, we aimed to pathophysiologically clarify the role of CD34 in HAVICs obtained from individual patients with severe CAVS. A DNA microarray between CD34-positive and CD34-negative HAVICs, separated by fluorescence-activated cell sorting, indicated that tenascin X (TNX) mRNA expression significantly decreased in CD34-negative cells. Furthermore, the inflammatory cytokines, tumor necrosis factor (TNF)-α and interleukin (IL)-1β significantly downregulated CD34 expression in HAVICs. TGF-β, a key cytokine of endothelial-mesenchymal transition, did not affect HAVIC calcification. CD34 overexpression strongly inhibited TNF-α- and IL-1β-induced calcification and maintained TNX mRNA expression. These results suggest one possibility that CD34 is an inhibitory regulator of valve calcification. Furthermore, TNF-α- and IL-1β-induced CD34 downregulation in HAVICs contributes to HAVIC calcification by downregulating TNX protein expression.
Collapse
Affiliation(s)
- Shihu Men
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan
| | - Zaiqiang Yu
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan.
| | - Xu Liu
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan
| | - Kazuyuki Daitoku
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan
| | - Mayuki Tachizaki
- Department of Vascular and Inflammatory Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan
| | - Shogo Kawaguchi
- Department of Vascular and Inflammatory Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan
| | - Tadaatsu Imaizumi
- Department of Vascular and Inflammatory Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan
| | - Masahito Minakawa
- Department of Thoracic and Cardiovascular Surgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan
| | - Kazuhiko Seya
- Department of Vascular and Inflammatory Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, 036-8562, Japan.
| |
Collapse
|
2
|
Bartoli-Leonard F, Pennel T, Caputo M. Immunotherapy in the Context of Aortic Valve Diseases. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07608-7. [PMID: 39017904 DOI: 10.1007/s10557-024-07608-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE Aortic valve disease (AVD) affects millions of people around the world, with no pharmacological intervention available. Widely considered a multi-faceted disease comprising both regurgitative pathogenesis, in which retrograde blood flows back through to the left ventricle, and aortic valve stenosis, which is characterized by the thickening, fibrosis, and subsequent mineralization of the aortic valve leaflets, limiting the anterograde flow through the valve, surgical intervention is still the main treatment, which incurs considerable risk to the patient. RESULTS Though originally thought of as a passive degeneration of the valve or a congenital malformation that has occurred before birth, the paradigm of AVD is shifting, and research into the inflammatory drivers of valve disease as a potential mechanism to modulate the pathobiology of this life-limiting pathology is taking center stage. Following limited success in mainstay therapeutics such as statins and mineralisation inhibitors, immunomodulatory strategies are being developed. Immune cell therapy has begun to be adopted in the cancer field, in which T cells (chimeric antigen receptor (CAR) T cells) are isolated from the patient, programmed to attack the cancer, and then re-administered to the patient. Within cardiac research, a novel T cell-based therapeutic approach has been developed to target lipid nanoparticles responsible for increasing cardiac fibrosis in a failing heart. With clonally expanded T-cell populations recently identified within the diseased valve, their unique epitope presentation may serve to identify novel targets for the treatment of valve disease. CONCLUSION Taken together, targeted T-cell therapy may hold promise as a therapeutic platform to target a multitude of diseases with an autoimmune aspect, and this review aims to frame this in the context of cardiovascular disease, delineating what is currently known in the field, both clinically and translationally.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK.
- Bristol Heart Institute, University Hospital Bristol and Weston NHS Foundation Trust, Bristol, UK.
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Cape Town, South Africa.
| | - Tim Pennel
- Chris Barnard Division of Cardiothoracic Surgery, University of Cape Town, Cape Town, South Africa
| | - Massimo Caputo
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, UK
- Bristol Heart Institute, University Hospital Bristol and Weston NHS Foundation Trust, Bristol, UK
| |
Collapse
|
3
|
Chen SY, Kong XQ, Zhang JJ. Pathological Mechanism and Treatment of Calcified Aortic Stenosis. Cardiol Rev 2024; 32:320-327. [PMID: 38848535 DOI: 10.1097/crd.0000000000000510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Calcified aortic stenosis (AS) is one of the most common valvular heart diseases worldwide, characterized by progressive fibrocalcific remodeling and thickening of the leaflets, which ultimately leads to obstruction of blood flow. Its pathobiology is an active and complicated process, involving endothelial cell dysfunction, lipoprotein deposition and oxidation, chronic inflammation, phenotypic transformation of valve interstitial cells, neovascularization, and intravalvular hemorrhage. To date, no targeted drug has been proven to slow down or prevent disease progression. Aortic valve replacement is still the optimal treatment of AS. This article reviews the etiology, diagnosis, and management of calcified aortic stenosis and proposes novel potential therapeutic targets.
Collapse
Affiliation(s)
- Si-Yu Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing 210006, China
| | - Xiang-Quan Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing 210006, China
- Department of Cardiology, Nanjing Heart Centre, Nanjing, China
| | - Jun-Jie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing 210006, China
- Department of Cardiology, Nanjing Heart Centre, Nanjing, China
| |
Collapse
|
4
|
Yang X, Zeng J, Xie K, Su S, Guo Y, Zhang H, Chen J, Ma Z, Xiao Z, Zhu P, Zheng S, Xu D, Zeng Q. Advanced glycation end product-modified low-density lipoprotein promotes pro-osteogenic reprogramming via RAGE/NF-κB pathway and exaggerates aortic valve calcification in hamsters. Mol Med 2024; 30:76. [PMID: 38840067 PMCID: PMC11155186 DOI: 10.1186/s10020-024-00833-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/09/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Advanced glycation end product-modified low-density lipoprotein (AGE-LDL) is related to inflammation and the development of atherosclerosis. Additionally, it has been demonstrated that receptor for advanced glycation end products (RAGE) has a role in the condition known as calcific aortic valve disease (CAVD). Here, we hypothesized that the AGE-LDL/RAGE axis could also be involved in the pathophysiological mechanism of CAVD. METHODS Human aortic valve interstitial cells (HAVICs) were stimulated with AGE-LDL following pre-treatment with or without interleukin 37 (IL-37). Low-density lipoprotein receptor deletion (Ldlr-/-) hamsters were randomly allocated to chow diet (CD) group and high carbohydrate and high fat diet (HCHFD) group. RESULTS AGE-LDL levels were significantly elevated in patients with CAVD and in a hamster model of aortic valve calcification. Our in vitro data further demonstrated that AGE-LDL augmented the expression of intercellular cell adhesion molecule-1 (ICAM-1), interleukin-6 (IL-6) and alkaline phosphatase (ALP) in a dose-dependent manner through NF-κB activation, which was attenuated by nuclear factor kappa-B (NF-κB) inhibitor Bay11-7082. The expression of RAGE was augmented in calcified aortic valves, and knockdown of RAGE in HAVICs attenuated the AGE-LDL-induced inflammatory and osteogenic responses as well as NF-κB activation. IL-37 suppressed inflammatory and osteogenic responses and NF-κB activation in HAVICs. The vivo experiment also demonstrate that supplementation with IL-37 inhibited valvular inflammatory response and thereby suppressed valvular osteogenic activities. CONCLUSIONS AGE-LDL promoted inflammatory responses and osteogenic differentiation through RAGE/NF-κB pathway in vitro and aortic valve lesions in vivo. IL-37 suppressed the AGE-LDL-induced inflammatory and osteogenic responses in vitro and attenuated aortic valve lesions in a hamster model of CAVD.
Collapse
Affiliation(s)
- Xi Yang
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Jingxin Zeng
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Kaiji Xie
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Shuwen Su
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Yuyang Guo
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Hao Zhang
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Jun Chen
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Zhuang Ma
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Peng Zhu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dingli Xu
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
| | - Qingchun Zeng
- State Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, 510515, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
| |
Collapse
|
5
|
Lin R, Zhu Y, Chen W, Wang Z, Wang Y, Du J. Identification of Circulating Inflammatory Proteins Associated with Calcific Aortic Valve Stenosis by Multiplex Analysis. Cardiovasc Toxicol 2024; 24:499-512. [PMID: 38589550 DOI: 10.1007/s12012-024-09854-5] [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: 03/06/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024]
Abstract
Calcific aortic valve stenosis (CAVS) is characterized by increasing inflammation and progressive calcification in the aortic valve leaflets and is a major cause of death in the aging population. This study aimed to identify the inflammatory proteins involved in CAVS and provide potential therapeutic targets. We investigated the observational and causal associations of 92 inflammatory proteins, which were measured using affinity-based proteomic assays. Firstly, the case-control cohort identified differential proteins associated with the occurrence and progression of CAVS. Subsequently, we delved into exploring the causal impacts of these associated proteins through Mendelian randomization. This involved utilizing genetic instruments derived from cis-protein quantitative loci identified in genome-wide association studies, encompassing a cohort of over 400,000 individuals. Finally, we investigated the gene transcription and protein expression levels of inflammatory proteins by single-cell and immunohistochemistry analysis. Multivariate logistic regression and spearman's correlation analysis showed that five proteins showed a significant positive correlation with disease severity. Mendelian randomization showed that elevated levels of two proteins, namely, matrix metallopeptidase-1 (MMP1) and sirtuin 2 (SIRT2), were associated with an increased risk of CAVS. Immunohistochemistry and single-cell transcriptomes showed that expression levels of MMP1 and SIRT2 at the tissue and cell levels were significantly higher in calcified valves than in non-calcified control valves. These findings indicate that MMP1 and SIRT2 are causally related to CAVS and open up the possibility for identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Rui Lin
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Yuexin Zhu
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Weiyao Chen
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Zhiao Wang
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Yuan Wang
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China.
| | - Jie Du
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
6
|
Liu X, Li T, Sun J, Wang Z. The Role of Endoplasmic Reticulum Stress in Calcific Aortic Valve Disease. Can J Cardiol 2023; 39:1571-1580. [PMID: 37516250 DOI: 10.1016/j.cjca.2023.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/28/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023] Open
Abstract
Calcific aortic valve disease (CAVD), which is involved in osteogenic reprogramming of valvular interstitial cells, is the most common form of valve disease. It still lacks effective pharmacologic intervention, as its cellular biological mechanisms remain unclear. Congenital abnormality (bicuspid valve) and older age are considered to be the most powerful risk factors for CAVD. Aortic valve sclerosis (AVS) and calcific aortic stenosis (CAS), 2 subclinical forms of CAVD, represent 2 distinct stages of aortic valve calcification. During the AVS stage, the disease is characterised by endothelial activation/damage, inflammatory response, and lipid infiltration accompanied by microcalcification. The CAS stage is dominated by calcification, resulting in valvular dysfunction and severe obstruction to cardiac outflow, which is life threatening if surgery is not performed in time. Endoplasmic reticulum (ER) stress, a state in which conditions disrupting ER homeostasis cause an accumulation of unfolded and misfolded proteins in the ER lumen, has been shown to promote osteogenic differentiation and aortic valve calcification. Therefore, identifying targets or drugs for suppressing ER stress may be a novel approach for CAVD treatment.
Collapse
Affiliation(s)
- Xiaolin Liu
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Medicial Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong, China
| | - Ting Li
- School of Life Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Jun Sun
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhengjun Wang
- Department of Cardiac Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
7
|
Ma W, Zhang W, Liu H, Qian B, Lai R, Yao Z, Wang Y, Yan Y, Yuan Z. Plasma Cell-Free DNA Is a Potential Biomarker for Diagnosis of Calcific Aortic Valve Disease. Cardiology 2023; 149:155-162. [PMID: 37899036 PMCID: PMC10994581 DOI: 10.1159/000534229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/14/2023] [Indexed: 10/31/2023]
Abstract
INTRODUCTION Calcific aortic valve disease (CAVD) is the third most common cardiovascular disease in aging populations. Despite a growing number of biomarkers having been shown to be associated with CAVD, a marker suitable for routine testing in clinical practice is still needed. Plasma cell-free DNA (cfDNA) has been suggested as a biomarker for diagnosis and prognosis in multiple diseases. In this study, we aimed to test whether cfDNA could be used as a biomarker for the diagnosis of CAVD. METHODS Serum samples were collected from 137 diagnosed CAVD patients and 180 normal controls. The amount of cfDNA was quantified by amplifying a short fragment (ALU 115) and a long fragment (ALU 247) using quantitative real-time PCR. The cfDNA integrity (cfDI) was calculated as the ratio of ALU247 to ALU115. The association between CAVD and cfDI was evaluated using regression analysis. RESULTS CAVD patients had increased ALU 115 fragments (median, 185.14 (416.42) versus 302.83 (665.41), p < 0.05) but a decreased value of cfDI (mean, 0.50 ± 0.25 vs. 0.41 ± 0.26, p < 0.01) in their serum when compared to controls. This difference was more dramatic in non-rheumatic CAVD patients (p < 0.001) versus rheumatic CAVD patients (no significant difference). Similarly, CAVD patients with bicuspid aortic valve (BAV) (p < 0.01) showed a greater difference than non-BAV CAVD patients (p < 0.05). Linear regression and logistic regression showed that cfDI was independently and significantly associated with the presence of CAVD (95% CI, 0.096 to 0.773, p < 0.05). The ROC assay revealed that cfDI combined with clinical characteristics had a better diagnostic value than cfDI alone (AUC = 0.6191, p < 0.001). CONCLUSION cfDI may be a potential biomarker for diagnosis of CAVD.
Collapse
Affiliation(s)
- Wangge Ma
- Department of Cardiology, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Wei Zhang
- Department of General Practice, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huahua Liu
- Department of Cardiology, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Benheng Qian
- Department of Cardiology, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Rongguang Lai
- Department of Cardiology, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zijun Yao
- Department of Cardiology, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yidong Wang
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, The Institute of Cardiovascular Sciences, School of Basic Medical Sciences; Department of Cardiology, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yang Yan
- Department of Cardiovascular Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zuyi Yuan
- Department of Cardiology, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
8
|
Shamekhi J, Uehre C, Al-Kassou B, Weber M, Sugiura A, Wilde N, Mauri V, Veulemans V, Kelm M, Baldus S, Nickenig G, Zimmer S. Echocardiographic Progression of Calcific Aortic Valve Disease in Patients with Preexisting Aortic Valve Sclerosis. Rev Cardiovasc Med 2023; 24:293. [PMID: 39077564 PMCID: PMC11273110 DOI: 10.31083/j.rcm2410293] [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: 03/08/2023] [Revised: 05/12/2023] [Accepted: 06/20/2023] [Indexed: 07/31/2024] Open
Abstract
Background We aimed to evaluate echocardiographic parameters to predict calcific aortic valve disease (CAVD) progression. CAVD ranges from aortic valve sclerosis (ASc) with no functional impairment of the aortic valve to severe aortic stenosis (AS). It remains uncertain, which patients with ASc have a particularly high risk of developing AS. Methods We included a total of 153 patients with visual signs of ASc and peak flow velocity (Vmax) below 2.5 m/s at baseline echocardiography. Progression of CAVD to AS was defined as an increase in Vmax ≥ 2.5 m/s with a delta of ≥ 0.1 m/s; stable ASc was defined as Vmax below 2.5 m/s and a delta < 0.1 m/s. Finally, we compared clinical and echocardiographic parameters between these two groups. Results The mean age at baseline was 73.5 ( ± 8.2) years and 66.7% were of male sex. After a mean follow-up of 1463 days, 57 patients developed AS, while 96 patients remained in the ASc group. The AS group showed significantly more calcification (p < 0.001) and thickening (p < 0.001) of the aortic valve cusps at baseline, although hemodynamics showed no evidence of AS in both groups (ASc group: Vmax 1.6 ± 0.3 m/s versus AS group: Vmax 1.9 ± 0.3 m/s; p < 0.001). Advanced calcification (odds ratio [OR]: 4.8, 95% confidence interval [CI]: 1.5-15.9; p = 0.009) and a cusp thickness > 0.26 cm (OR: 16.6, 95% CI: 5.4-50.7; p < 0.001) were independent predictors for the development of AS. Conclusions The acquisition of simple echocardiographic parameter may help to identify patients with particularly high risk of developing AS.
Collapse
Affiliation(s)
- Jasmin Shamekhi
- Heart Center, Department of Medicine II, University Hospital Bonn, 53127
Bonn, Germany
| | - Carina Uehre
- Heart Center, Department of Medicine II, University Hospital Bonn, 53127
Bonn, Germany
| | - Baravan Al-Kassou
- Heart Center, Department of Medicine II, University Hospital Bonn, 53127
Bonn, Germany
| | - Marcel Weber
- Heart Center, Department of Medicine II, University Hospital Bonn, 53127
Bonn, Germany
| | - Atsushi Sugiura
- Heart Center, Department of Medicine II, University Hospital Bonn, 53127
Bonn, Germany
| | - Nihal Wilde
- Heart Center, Department of Medicine II, University Hospital Bonn, 53127
Bonn, Germany
| | - Victor Mauri
- Heart Center, Department of Cardiology, University Hospital Cologne, 50937
Cologne, Germany
| | - Verena Veulemans
- Heart Center, Department of Cardiology, University Hospital
Düsseldorf, 40225 Düsseldorf, Germany
| | - Malte Kelm
- Heart Center, Department of Cardiology, University Hospital
Düsseldorf, 40225 Düsseldorf, Germany
| | - Stephan Baldus
- Heart Center, Department of Cardiology, University Hospital Cologne, 50937
Cologne, Germany
| | - Georg Nickenig
- Heart Center, Department of Medicine II, University Hospital Bonn, 53127
Bonn, Germany
| | - Sebastian Zimmer
- Heart Center, Department of Medicine II, University Hospital Bonn, 53127
Bonn, Germany
| |
Collapse
|
9
|
Fang J, Qian Y, Chen J, Xu D, Cao N, Zhu G, Hu W, Hu H, Qian N, Yang S, Wang J, Liu X. Human antigen R regulates autophagic flux by stabilizing autophagy-associated mRNA in calcific aortic valve disease. Cardiovasc Res 2023; 119:2117-2129. [PMID: 37183487 DOI: 10.1093/cvr/cvad077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
AIMS The incidence of calcific aortic valve disease (CAVD) has risen over the last decade and is expected to continue rising; however, pharmacological approaches have proven ineffective. In this study, we evaluated the role and underlying mechanisms of human antigen R (HuR)-mediated post-transcriptional regulation in CAVD. METHODS AND RESULTS We found that HuR was significantly upregulated in human calcified aortic valves and primary aortic valvular interstitial cells (VICs) following osteogenic stimulation. Subsequent functional studies revealed that HuR silencing ameliorated calcification both in vitro and in vivo. For the first time, we demonstrated that HuR directly interacted with the transcript of phosphatidylinositol-5-phosphate 4-kinase, type II, alpha (PIP4K2A), which mediates phosphatidylinositol signalling, facilitates autophagy, and acts as an mRNA stabilizer. HuR positively modulated PIP4K2A expression at the post-transcriptional level and consequently influenced the AKT/mTOR/ATG13 pathway to regulate autophagy and CAVD progression. CONCLUSION Our study provides new insights into the post-transcriptional regulatory role of HuR in modulating autophagy-positive factors to regulate the pathogenesis of CAVD. Our findings highlight the potential of HuR as an innovative therapeutic target in CAVD treatment.
Collapse
Affiliation(s)
- Juan Fang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yi Qian
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Department of Cardiology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, China
| | - Jinyong Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Dilin Xu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Naifang Cao
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Gangjie Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Wangxing Hu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Haochang Hu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ningjing Qian
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Shuangshuang Yang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jian'an Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xianbao Liu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
10
|
Liu X, Zheng Q, Wang K, Luo J, Wang Z, Li H, Liu Z, Dong N, Shi J. Sam68 promotes osteogenic differentiation of aortic valvular interstitial cells by TNF-α/STAT3/autophagy axis. J Cell Commun Signal 2023; 17:863-879. [PMID: 36847917 PMCID: PMC10409708 DOI: 10.1007/s12079-023-00733-2] [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: 10/10/2022] [Accepted: 02/10/2023] [Indexed: 03/01/2023] Open
Abstract
Calcified aortic valve disease (CAVD) is a major non-rheumatic heart valve disease in the world, with a high mortality rate and without suitable pharmaceutical therapy due to its complex mechanisms. Src-associated in mitosis 68-KD (Sam68), an RNA binding protein, has been reported as a signaling adaptor in numerous signaling pathways (Huot in Mol Cell Biol, 29(7), 1933-1943, 2009), particularly in inflammatory signaling pathways. The effects of Sam68 on the osteogenic differentiation process of hVICs and its regulation on signal transducer and activator of transcription 3 (STAT3) signaling pathway have been investigated in this study. Human aortic valve samples detection found that Sam68 expression was up-regulated in human calcific aortic valves. We used tumor necrosis factor α (TNF-α) as an activator for osteogenic differentiation in vitro and the result indicated that Sam68 was highly expressed after TNF-α stimulation. Overexpression of Sam68 promoted osteogenic differentiation of hVICs while Sam68 knockdown reversed this effect. Sam68 interaction with STAT3 was predicted by using String database and was verified in this study. Sam68 knockdown reduced phosphorylation of STAT3 activated by TNF-α and the downstream gene expression, which further influenced autophagy flux in hVICs. STAT3 knockdown alleviated the osteogenic differentiation and calcium deposition promoted by Sam68 overexpression. In conclusion, Sam68 interacts with STAT3 and participates in its phosphorylation to promote osteogenic differentiation of hVICs to induce valve calcification. Thus, Sam68 may be a new therapeutic target for CAVD. Regulatory of Sam68 in TNF-α/STAT3/Autophagy Axis in promoting osteogenesis of hVICs.
Collapse
Affiliation(s)
- Xing Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| | - Qiang Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| | - Kan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| | - Jinjing Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| | - Zhijie Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| | - Huadong Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| | - Zongtao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022 People’s Republic of China
| |
Collapse
|
11
|
Chen J, Lyu L, Shen J, Pan Y, Jing J, Wang YJ, Wei T. Epidemiological study of calcified aortic valve stenosis in a Chinese community population. Postgrad Med J 2023; 99:868-874. [PMID: 37117153 DOI: 10.1136/pmj-2022-141721] [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: 03/17/2022] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Due to the ageing global population, calcified aortic valve disease is currently the most common cardiac valve disorder. This study aimed to investigate the prevalence and the risk factors for calcified aortic valve stenosis (CAVS), and develop a prediction model for predicting CAVS risk. METHODS AND RESULTS This study was derived from the cross-sectional baseline survey of the PRECISE study (NCT03178448). The demographic, clinical and laboratory information of each participant was obtained. Univariable and multivariable logistic regression models were used to determine CAVS risk factors. A prediction model for predicting CAVS risk based on risk factors was developed and the result was performed by nomogram. The discrimination of the prediction model was assessed by receiver operating characteristic curve analysis. The degree of fitting for the prediction model was assessed by calibration curve analysis. A total of 3067 participants (1427 men and 1640 women) were included. The prevalence of CAVS among those aged below 60 years old, 60-70 years old and over 70 years old was 4.1%, 10.3% and 21.9%, respectively. Multivariable regression analysis revealed that age (OR: 1.099; 95% CI: 1.076 to 1.123, p<0.001), pulse pressure (OR: 1.020; 95% CI: 1.009 to 1.031, p<0.001), uric acid (OR: 1.003; 95% CI: 1.001 to 1.004, p<0.001), glycosylated haemoglobin (HbA1c) (OR: 1.152; 95% CI: 1.028 to 1.292, p=0.015) and lipoprotein(a) (OR: 1.002; 95% CI: 1.001 to 1.002, p<0.001) were independent risk factors for CAVS. High-density lipoprotein cholesterol (HDL-C) was a protective factor for CAVS (OR: 0.539; 95% CI: 0.349 to 0.831, p=0.005). The prediction model including the above risk factors showed a risk prediction of CAVS with good discrimination. The area under the curve value was found to be 0.743 (95% CI: 0.711 to 0.775). CONCLUSION CAVS is currently prevalent in the elderly Chinese population. Age, pulse pressure, HbA1c, lower-level HDL-C, lipoprotein(a) and uric acid are the independent risk factors for CAVS.
Collapse
Affiliation(s)
- Jun Chen
- Department of Cardiology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, Zhejiang, China
- Department of Cardiology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lingchun Lyu
- Department of Cardiology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, Zhejiang, China
| | - Jiayi Shen
- Department of Cardiology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, Zhejiang, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Beijing, China
| | - Jing Jing
- Department of Neurology, Beijing Tiantan Hospital, Beijing, China
| | - Yong-Jun Wang
- Department of Neurology, Beijing Tiantan Hospital, Beijing, China
| | - Tiemin Wei
- Department of Cardiology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, Zhejiang, China
| |
Collapse
|
12
|
Zhu X, Yang L, Han X, Huang C, Huang G, Wei T, Shu L, Xu J. Oxidized phospholipids facilitate calcific aortic valve disease by elevating ATF4 through the PERK/eIF2α axis. Aging (Albany NY) 2023; 15:6834-6847. [PMID: 37462732 PMCID: PMC10415544 DOI: 10.18632/aging.204875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/15/2023] [Indexed: 08/07/2023]
Abstract
In this study we sought to analyze the critical role of oxidized phospholipid (OxPL) in the progression of calcific aortic valve disease (CAVD) with the involvement of activating transcription factor 4 (ATF4). Differentially expressed genes related to CAVD were identified using bioinformatics analysis. Expression of ATF4 was examined in mouse models of aortic valve calcification (AVC) induced by the high cholesterol (HC) diet. Valvular interstitial cells (VICs) were then isolated from mouse non-calcified valve tissues, induced by osteogenic induction medium (OIM) and co-cultured with OxPAPC-stimulated macrophages. The effect of OxPLs regulating ATF4 on the macrophage polarization and osteogenic differentiation of VICs was examined with gain- and loss-of-function experiments in VICs and in vivo. In aortic valve tissues and OIM-induced VICs, ATF4 was highly expressed. ATF4 knockdown alleviated the osteogenic differentiation of VICs, as evidenced by reduced expression of bone morphogenetic protein-2 (BMP2), osteopontin (OPN), and osteocalcin. In addition, knockdown of ATF4 arrested the AVC in vivo. Meanwhile, OxPL promoted M1 polarization of macrophages and mediated osteogenic differentiation of VICs. Furthermore, OxPL up-regulated ATF4 expression through protein kinase R-like endoplasmic reticulum kinase (PERK)/eukaryotic translation initiation factor 2 subunit alpha (eIF2α) pathway. In conclusion, OxPL can potentially up-regulate the expression of ATF4, inducing macrophages polarized to M1 phenotype, osteogenic differentiation of VICs and AVC, thus accelerating the progression of CAVD.
Collapse
Affiliation(s)
- Xiaohua Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Linjie Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Xu Han
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Chen Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Gongcheng Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Tingju Wei
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Liliang Shu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Jing Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| |
Collapse
|
13
|
Leibowitz D, Yoshida Y, Jin Z, Nakanishi K, Mannina C, Elkind MSV, Rundek T, Homma S, Sacco RL, Di Tullio MR. Factors associated with the progression of aortic valve calcification in older adults. Int J Cardiol 2023; 381:76-80. [PMID: 37030403 PMCID: PMC10161393 DOI: 10.1016/j.ijcard.2023.03.059] [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: 12/06/2022] [Revised: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND Aortic valve calcification (AVC) is a common valvular abnormality that predisposes to stenosis; AVC progression and factors associated with it remain unclear. We investigated the association of clinical factors and serum biomarkers with AVC progression in a population-based cohort of older adults. METHODS Participants enrolled in both the Cardiovascular Abnormalities and Brain Lesion study (CABL; years 2005-2010) and the Subclinical Atrial Fibrillation And Risk of Ischemic Stroke study (SAFARIS;2014-2019) represent the study cohort. AVC was defined as bright dense echoes >1 mm in size on ≥1 cusps; each cusp was graded on a scale of 0 (normal) to 3 (severe calcification) at baseline and follow up. Serum biomarkers were measured at the time of follow-up assessment. RESULTS 373 participants (mean 68.1 ± 7.6 years of age, 146 M/ 227F) were included. 139 (37%) had AVC progression;93 (25%) had mild progression (1 grade), and 46 (12%) had moderate-severe progression (≥2 grades). The only significant clinical predictor of any progression was the use of anti-hypertensive medication which was associated with older age, higher BMI and more frequent hypertension, diabetes and hyperlipidemia. In multivariable analysis including biomarkers, transforming growth factor beta 1 (TGF-β1) was significantly associated with both all and moderate-severe AVC progression. CONCLUSIONS A significant number of elderly subjects with AVC show progression of their valve disease; individual vascular risk factors are not associated with AVC progression, although a combined effect may exist. Higher levels of TGF-β1 are observed in individuals with AVC progression.
Collapse
Affiliation(s)
- David Leibowitz
- Heart Institute, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuriko Yoshida
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Zhezhen Jin
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Koki Nakanishi
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
| | - Carlo Mannina
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Mitchell S V Elkind
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Tatjana Rundek
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, FL, USA; Department of Public Health Sciences, Miller School of Medicine, University of Miami, FL, USA; Clinical and Translational Science Institute, Miller School of Medicine, University of Miami, FL, USA
| | - Shunichi Homma
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ralph L Sacco
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, FL, USA; Department of Public Health Sciences, Miller School of Medicine, University of Miami, FL, USA; Clinical and Translational Science Institute, Miller School of Medicine, University of Miami, FL, USA
| | - Marco R Di Tullio
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
14
|
Iqbal F, Schlotter F, Becker-Greene D, Lupieri A, Goettsch C, Hutcheson JD, Rogers MA, Itoh S, Halu A, Lee LH, Blaser MC, Mlynarchik AK, Hagita S, Kuraoka S, Chen HY, Engert JC, Passos LSA, Jha PK, Osborn EA, Jaffer FA, Body SC, Robson SC, Thanassoulis G, Aikawa M, Singh SA, Sonawane AR, Aikawa E. Sortilin enhances fibrosis and calcification in aortic valve disease by inducing interstitial cell heterogeneity. Eur Heart J 2023; 44:885-898. [PMID: 36660854 PMCID: PMC9991042 DOI: 10.1093/eurheartj/ehac818] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 11/29/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023] Open
Abstract
AIMS Calcific aortic valve disease (CAVD) is the most common valve disease, which consists of a chronic interplay of inflammation, fibrosis, and calcification. In this study, sortilin (SORT1) was identified as a novel key player in the pathophysiology of CAVD, and its role in the transformation of valvular interstitial cells (VICs) into pathological phenotypes is explored. METHODS AND RESULTS An aortic valve (AV) wire injury (AVWI) mouse model with sortilin deficiency was used to determine the effects of sortilin on AV stenosis, fibrosis, and calcification. In vitro experiments employed human primary VICs cultured in osteogenic conditions for 7, 14, and 21 days; and processed for imaging, proteomics, and transcriptomics including single-cell RNA-sequencing (scRNA-seq). The AVWI mouse model showed reduced AV fibrosis, calcification, and stenosis in sortilin-deficient mice vs. littermate controls. Protein studies identified the transition of human VICs into a myofibroblast-like phenotype mediated by sortilin. Sortilin loss-of-function decreased in vitro VIC calcification. ScRNA-seq identified 12 differentially expressed cell clusters in human VIC samples, where a novel combined inflammatory myofibroblastic-osteogenic VIC (IMO-VIC) phenotype was detected with increased expression of SORT1, COL1A1, WNT5A, IL-6, and serum amyloid A1. VICs sequenced with sortilin deficiency showed decreased IMO-VIC phenotype. CONCLUSION Sortilin promotes CAVD by mediating valvular fibrosis and calcification, and a newly identified phenotype (IMO-VIC). This is the first study to examine the role of sortilin in valvular calcification and it may render it a therapeutic target to inhibit IMO-VIC emergence by simultaneously reducing inflammation, fibrosis, and calcification, the three key pathological processes underlying CAVD.
Collapse
Affiliation(s)
- Farwah Iqbal
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Florian Schlotter
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Cardiology, Heart Center Leipzig at Leipzig University, Leipzig, Germany
| | - Dakota Becker-Greene
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Adrien Lupieri
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Claudia Goettsch
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA
| | - Maximillian A Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Shinsuke Itoh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Arda Halu
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lang Ho Lee
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew K Mlynarchik
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sumihiko Hagita
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Shiori Kuraoka
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hao Yu Chen
- Department of Medicine, McGill University, Montreal, Canada
| | - James C Engert
- Department of Medicine, McGill University, Montreal, Canada
| | - Livia S A Passos
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Prabhash K Jha
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric A Osborn
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Farouc A Jaffer
- Cardiovascular Research Center, Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon C Body
- Department of Anesthesiology, Boston University School of Medicine, Boston, MA, USA
| | - Simon C Robson
- Center for Inflammation Research, Department of Anesthesia, BIDMC, Harvard Medical School, Boston, MA, USA
| | | | - Masanori Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Abhijeet R Sonawane
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Kologrivova IV, Naryzhnaya NV, Koshelskaya OA, Suslova TE, Kravchenko ES, Kharitonova OA, Evtushenko VV, Boshchenko AA. Association of Epicardial Adipose Tissue Adipocytes Hypertrophy with Biomarkers of Low-Grade Inflammation and Extracellular Matrix Remodeling in Patients with Coronary Artery Disease. Biomedicines 2023; 11:biomedicines11020241. [PMID: 36830779 PMCID: PMC9953115 DOI: 10.3390/biomedicines11020241] [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: 12/23/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
The aim of the study was to compare the morphological features of epicardial adipose tissue (EAT) adipocyte with the circulating inflammatory biomarkers and parameters of extracellular matrix remodeling in patients with coronary artery disease (CAD). We recruited 42 patients with CAD (m/f 28/14) who were scheduled for coronary artery bypass graft surgery (CABG). EAT adipocytes were obtained by the enzymatic method from intraoperative adipose tissue samples. Concentrations of secreted and lipoprotein-associated phospholipase A2 (sPLA2 and LpPLA2), TNF-α, IL-1β, IL-6, IL-10, high-sensitive C-reactive protein (hsCRP), metalloproteinase-9 (MMP-9), MMP-2, C-terminal cross-linking telopeptide of type I collagen (CTX-I), and tissue inhibitor of metalloproteinase 1 (TIMP-1) were measured in blood serum. Patients were divided into two groups: group 1-with mean EAT adipocytes' size ≤ 87.32 μm; group 2-with mean EAT adipocytes' size > 87.32 μm. Patients of group 2 had higher concentrations of triglycerides, hsCRP, TNF-α, and sPLA2 and a lower concentration of CTX-I. A multiple logistic regression model was created (RN2 = 0.43, p = 0.0013). Concentrations of TNF-α, sPLA2 and CTX-I appeared to be independent determinants of the EAT adipocyte hypertrophy. ROC analysis revealed the 78% accuracy, 71% sensitivity, and 85% specificity of the model, AUC = 0.82. According to our results, chronic low-grade inflammation and extracellular matrix remodeling are closely associated with the development of hypertrophy of EAT adipocytes, with serum concentrations of TNF-α, sPLA2 and CTX-I being the key predictors, describing the variability of epicardial adipocytes' size.
Collapse
Affiliation(s)
- Irina V. Kologrivova
- Correspondence: (I.V.K.); (N.V.N.); Tel.: +79-131-053-869 (I.V.K.); +79-039-542-139 (N.V.N.)
| | - Natalia V. Naryzhnaya
- Correspondence: (I.V.K.); (N.V.N.); Tel.: +79-131-053-869 (I.V.K.); +79-039-542-139 (N.V.N.)
| | | | | | | | | | | | | |
Collapse
|
16
|
Marreiros C, Viegas C, Simes D. Targeting a Silent Disease: Vascular Calcification in Chronic Kidney Disease. Int J Mol Sci 2022; 23:16114. [PMID: 36555758 PMCID: PMC9781141 DOI: 10.3390/ijms232416114] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Chronic kidney disease (CKD) patients have a higher risk of developing early cardiovascular disease (CVD). Although vascular calcification (VC) is one of the strongest predictors of CVD risk, its diagnosis among the CKD population remains a serious clinical challenge. This is mainly due to the complexity of VC, which results from various interconnected pathological mechanisms occurring at early stages and at multiples sites, affecting the medial and intimal layers of the vascular tree. Here, we review the most used and recently developed imaging techniques, here referred to as imaging biomarkers, for VC detection and monitoring, while discussing their strengths and limitations considering the specificities of VC in a CKD context. Although imaging biomarkers have a crucial role in the diagnosis of VC, with important insights into CVD risk, circulating biomarkers represent an added value by reflecting the molecular dynamics and mechanisms involved in VC pathophysiological pathways, opening new avenues into the early detection and targeted interventions. We propose that a combined strategy using imaging and circulating biomarkers with a role in multiple VC molecular mechanisms, such as Fetuin-A, Matrix Gla protein, Gla-rich protein and calciprotein particles, should represent high prognostic value for management of CVD risk in the CKD population.
Collapse
Affiliation(s)
- Catarina Marreiros
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Carla Viegas
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Dina Simes
- Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
17
|
Lv X, Wang X, Liu J, Wang F, Sun M, Fan X, Ye Z, Liu P, Wen J. Potential biomarkers and immune cell infiltration involved in aortic valve calcification identified through integrated bioinformatics analysis. Front Physiol 2022; 13:944551. [PMID: 36589450 PMCID: PMC9797982 DOI: 10.3389/fphys.2022.944551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Calcific aortic valve disease (CAVD) is the most common valvular heart disease in the aging population, resulting in a significant health and economic burden worldwide, but its underlying diagnostic biomarkers and pathophysiological mechanisms are not fully understood. Methods: Three publicly available gene expression profiles (GSE12644, GSE51472, and GSE77287) from human Calcific aortic valve disease (CAVD) and normal aortic valve samples were downloaded from the Gene Expression Omnibus database for combined analysis. R software was used to identify differentially expressed genes (DEGs) and conduct functional investigations. Two machine learning algorithms, least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE), were applied to identify key feature genes as potential biomarkers for Calcific aortic valve disease (CAVD). Receiver operating characteristic (ROC) curves were used to evaluate the discriminatory ability of key genes. The CIBERSORT deconvolution algorithm was used to determine differential immune cell infiltration and the relationship between key genes and immune cell types. Finally, the Expression level and diagnostic ability of the identified biomarkers were further validated in an external dataset (GSE83453), a single-cell sequencing dataset (SRP222100), and immunohistochemical staining of human clinical tissue samples, respectively. Results: In total, 34 identified DEGs included 21 upregulated and 13 downregulated genes. DEGs were mainly involved in immune-related pathways such as leukocyte migration, granulocyte chemotaxis, cytokine activity, and IL-17 signaling. The machine learning algorithm identified SCG2 and CCL19 as key feature genes [area under the ROC curve (AUC) = 0.940 and 0.913, respectively; validation AUC = 0.917 and 0.903, respectively]. CIBERSORT analysis indicated that the proportion of immune cells in Calcific aortic valve disease (CAVD) was different from that in normal aortic valve tissues, specifically M2 and M0 macrophages. Key genes SCG2 and CCL19 were significantly positively correlated with M0 macrophages. Single-cell sequencing analysis and immunohistochemical staining of human aortic valve tissue samples showed that SCG2 and CCL19 were increased in Calcific aortic valve disease (CAVD) valves. Conclusion: SCG2 and CCL19 are potential novel biomarkers of Calcific aortic valve disease (CAVD) and may play important roles in the biological process of Calcific aortic valve disease (CAVD). Our findings advance understanding of the underlying mechanisms of Calcific aortic valve disease (CAVD) pathogenesis and provide valuable information for future research into novel diagnostic and immunotherapeutic targets for Calcific aortic valve disease (CAVD).
Collapse
Affiliation(s)
- Xiaoshuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China,Graduate School of Peking Union Medical College, Beijing, China
| | - Xiaohui Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China,Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Jingwen Liu
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China,Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Feng Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China,Graduate School of Peking Union Medical College, Beijing, China
| | - Mingsheng Sun
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China,Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xueqiang Fan
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Peng Liu
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianyan Wen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China,*Correspondence: Jianyan Wen,
| |
Collapse
|
18
|
[Optimal time window for observation of calcific aortic valve disease in mice following catheter-induced valve injury]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1532-1538. [PMID: 36329588 PMCID: PMC9637488 DOI: 10.12122/j.issn.1673-4254.2022.10.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To investigate the optimal time window for observation of catheter-induced valve injury that mimics calcified aortic valve disease in mice. METHODS A catheter was inserted into the right common carotid artery of 8-week-old C57BL6 mice under ultrasound guidance, and aortic valve injury was induced using the guide wire.At 4, 8 and 16 weeks after modeling, the mice were subjected to ultrasound measurement of the heart short axial shortening rate, aortic valve peak velocity and aortic valve orifice area.Grain-Eosin staining was used to observe the changes in the thickness of the aortic valve, and calcium deposition in the aortic valve was assessed using Alizarin red staining.Immunofluorescence assay was performed to detect the expression of alkaline phosphatase (ALP) in the aortic valve. RESULTS At 4, 8 and 16 weeks after modeling, valve thickness (P=0.002), calcium deposition (P < 0.0001) and the expression of osteogenic protein ALP (P=0.0016) were significantly increased, but their increments were comparable at the 3 time points of observation. CONCLUSION In mouse models of calcific aortic valve disease induced by catheter valve injury, 4 weeks after the injury appears to be the optimal time window for observation of pathophysiological changes in the aortic valves to avoid further increase of the death rate of the mice over time.
Collapse
|
19
|
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
|
20
|
Serum and Vascular Stiffness Biomarkers Associated with the Severity of Degenerative Aortic Valve Stenosis and Cardiovascular Outcomes. J Cardiovasc Dev Dis 2022; 9:jcdd9060193. [PMID: 35735822 PMCID: PMC9225443 DOI: 10.3390/jcdd9060193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/07/2022] [Accepted: 06/16/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Although degenerative aortic valve stenosis (DAS) is the most prevalent growth-up congestive heart valve disease, still little known about relationships between DAS severity, vascular stiffness (VS), echocardiographic parameters, and serum biomarkers in patients undergoing transcatheter (TAVR) or surgical aortic valve replacement (SAVR). The objective of this study was to identify biomarkers associated with DAS severity, and those that are associated with cardiovascular death (CVD) and episodes of chronic heart failure (CHF) exacerbation. Methods: A total of 137 patients with initially moderate-to-severe DAS were prospectively evaluated for the relationship between DAS severity, baseline VS, and serum biomarkers (uPAR, GDF-15, Gal-3, IL-6Rα, ET-1, PCSK9, RANTES/CCL5, NT-proBNP, and hs-TnT), and were followed-up for 48 months. The prognostic significance of each variable for CVD and CHF risk was measured by hazard ratio of risk (HR), which was calculated by Cox’s proportional hazard model. Results: DAS severity showed correlations with IL-6Rα (r = 0.306, p < 0.001), uPAR (r = 0.184, p = 0.032), and NT-proBNP (r = −0.389, p < 0.001). Levels of ET-1 and Gal-3 were strongly correlated with VS parameters (r = 0.674, p < 0.001; r = 0.724, p < 0.001). Out of 137 patients, 20 were referred to TAVR, 88 to SAVR, and 29 to OMT. In TAVR patients, the highest levels of ET-1, Gal-3, and VS were found as compared to other patients. The highest incidence of CVD was observed in patients who underwent TAVR (35%), compared to SAVR (8%) and OMT (10.3%) (p = 0.004). In a multivariate analysis, ET-1 occurred predictive of CVD risk (HR 25.1, p = 0.047), while Gal-3 > 11.5 ng/mL increased the risk of CHF exacerbation episodes requiring hospital admission by 12%. Conclusions: Our study indicated that ET-1 and Gal-3 levels may be associated with the outcomes in patients with DAS.
Collapse
|
21
|
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
|
22
|
Liu Q, Yu Y, Xi R, Li J, Lai R, Wang T, Fan Y, Zhang Z, Xu H, Ju J. Association Between Lipoprotein(a) and Calcific Aortic Valve Disease: A Systematic Review and Meta-Analysis. Front Cardiovasc Med 2022; 9:877140. [PMID: 35548407 PMCID: PMC9082602 DOI: 10.3389/fcvm.2022.877140] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/28/2022] [Indexed: 01/03/2023] Open
Abstract
Background Preliminary studies indicated that enhanced plasma levels of lipoprotein(a) [lp(a)] might link with the risk of calcific aortic valve disease (CAVD), but the clinical association between them remained inconclusive. This systematic review and meta-analysis were aimed to determine this association. Methods We comprehensively searched PubMed, Embase, Web of Science, and Scopus databases for studies reporting the incidence of CAVD and their plasma lp(a) concentrations. Pooled risk ratio (RR) and 95% confidence interval (95% CI) were calculated to evaluate the effect of lp(a) on CAVD using the random-effects model. Subgroup analyses by study types, countries, and the level of adjustment were also conducted. Funnel plots, Egger's test and Begg's test were conducted to evaluate the publication bias. Results Eight eligible studies with 52,931 participants were included in this systematic review and meta-analysis. Of these, four were cohort studies and four were case-control studies. Five studies were rated as high quality, three as moderate quality. The pooled results showed that plasma lp(a) levels ≥50 mg/dL were associated with a 1.76-fold increased risk of CAVD (RR, 1.76; 95% CI, 1.47–2.11), but lp(a) levels ≥30 mg/dL were not observed to be significantly related with CAVD (RR, 1.28; 95% CI, 0.98–1.68). We performed subgroup analyses by study type, the RRs of cohort studies revealed lp(a) levels ≥50 mg/dL and lp(a) levels ≥30 mg/dL have positive association with CAVD (RR, 1.70; 95% CI, 1.39–2.07; RR 1.38; 95% CI, 1.19–1.61). Conclusion High plasma lp(a) levels (≥50 mg/dL) are significantly associated with increased risk of CAVD.
Collapse
Affiliation(s)
- Qiyu Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yanqiao Yu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ruixi Xi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingen Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Runmin Lai
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongxin Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yixuan Fan
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zihao Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Hao Xu
| | - Jianqing Ju
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Jianqing Ju
| |
Collapse
|
23
|
Prioritization of Candidate Biomarkers for Degenerative Aortic Stenosis through a Systems Biology-Based In-Silico Approach. J Pers Med 2022; 12:jpm12040642. [PMID: 35455758 PMCID: PMC9026876 DOI: 10.3390/jpm12040642] [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: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Degenerative aortic stenosis is the most common valve disease in the elderly and is usually confirmed at an advanced stage when the only treatment is surgery. This work is focused on the study of previously defined biomarkers through systems biology and artificial neuronal networks to understand their potential role within aortic stenosis. The goal was generating a molecular panel of biomarkers to ensure an accurate diagnosis, risk stratification, and follow-up of aortic stenosis patients. We used in silico studies to combine and re-analyze the results of our previous studies and, with information from multiple databases, established a mathematical model. After this, we prioritized two proteins related to endoplasmic reticulum stress, thrombospondin-1 and endoplasmin, which have not been previously validated as markers for aortic stenosis, and analyzed them in a cell model and in plasma from human subjects. Large-scale bioinformatics tools allow us to extract the most significant results after using high throughput analytical techniques. Our results could help to prevent the development of aortic stenosis and open the possibility of a future strategy based on more specific therapies.
Collapse
|
24
|
Ferrari S, Pesce M. The Complex Interplay of Inflammation, Metabolism, Epigenetics, and Sex in Calcific Disease of the Aortic Valve. Front Cardiovasc Med 2022; 8:791646. [PMID: 35071359 PMCID: PMC8770423 DOI: 10.3389/fcvm.2021.791646] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022] Open
Abstract
Calcification of the aortic valve is one of the most rapidly increasing pathologies in the aging population worldwide. Traditionally associated to cardiovascular risk conditions, this pathology is still relatively unaddressed on a molecular/cellular standpoint and there are no available treatments to retard its progression unless valve substitution. In this review, we will describe some of the most involved inflammatory players, the metabolic changes that may be responsible of epigenetic modifications and the gender-related differences in the onset of the disease. A better understanding of these aspects and their integration into a unique pathophysiology context is relevant to improve current therapies and patients management.
Collapse
Affiliation(s)
- Silvia Ferrari
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| |
Collapse
|
25
|
Exploring potential genes and pathways related to calcific aortic valve disease. Gene 2022; 808:145987. [PMID: 34600049 DOI: 10.1016/j.gene.2021.145987] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/10/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Calcific aortic valve disease (CAVD) is currently the most prevalent valvular disease. However, the pathological mechanism of CAVD has not yet been fully elucidated, and no drugs can delay or halt the progression of CAVD. This study aimed to screen for potential biomarkers and pathways of CAVD through bioinformatics analysis. The identification of differentially expressed genes (DEGs) between calcific aortic valves and the control group was performed based on four microarray datasets: GSE12644, GSE51472, GSE77287 and GSE83453. Gene Ontology and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment analysis were conducted. Furthermore, the protein-protein interaction network, and microRNA-target interaction was performed, and hub genes were obtained by using twelve cytoHubba algorithms. As a result, 327 DEGs were identified, including 206 up-regulated and 121 down-regulated genes. KEGG analysis showed that these DEGs were mainly enriched in the PI3K-AKT signaling pathway, ECM-receptor interaction, cytokine-cytokine receptor interaction, and chemokine signaling pathway etc. Moreover, we identified 19 hub genes: CXCL8, CXCL12, CSF1R, HCK, PLEK, CCL5, TLR8, VCAM1, CCR1, CCR7, FPR1, TYROBP, CX3CR1, KIT, PPBP, SPP1, SYK, TLR7, and VWF. And multiple potential miRNAs, including miR-141, miR-34a, miR-155, and miR-486, were identified. And western blot was performed to validate the expression level of hub genes. In conclusion, this study identified several promising biomarkers and pathways for CAVD, which may provide novel molecular markers for diagnosis and targeted therapy.
Collapse
|
26
|
Metformin alleviates the calcification of aortic valve interstitial cells through activating the PI3K/AKT pathway in an AMPK dependent way. Mol Med 2021; 27:156. [PMID: 34895136 PMCID: PMC8666063 DOI: 10.1186/s10020-021-00416-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is the most prevalent valvular disease worldwide. However, no effective treatment could delay or prevent the progression of the disease due to the poor understanding of its pathological mechanism. Many studies showed that metformin exerted beneficial effects on multiple cardiovascular diseases by mediating multiple proteins such as AMPK, NF-κB, and AKT. This study aims to verify whether metformin can inhibit aortic calcification through the PI3K/AKT signaling pathway. METHODS We first analyzed four microarray datasets to screen differentially expressed genes (DEGs) and signaling pathways related to CAVD. Then aortic valve samples were used to verify selected genes and pathways through immunohistochemistry (IHC) and western blot (WB) assays. Aortic valve interstitial cells (AVICs) were isolated from non-calcific aortic valves and then cultured with phosphate medium (PM) with or without metformin to verify whether metformin can inhibit the osteogenic differentiation and calcification of AVICs. Finally, we used inhibitors and siRNA targeting AMPK, NF-κB, and AKT to study the mechanism of metformin. RESULTS We screened 227 DEGs; NF-κB and PI3K/AKT signaling pathways were implicated in the pathological mechanism of CAVD. IHC and WB experiments showed decreased AMPK and AKT and increased Bax in calcific aortic valves. PM treatment significantly reduced AMPK and PI3K/AKT signaling pathways, promoted Bax/Bcl2 ratio, and induced AVICs calcification. Metformin treatment ameliorated AVICs calcification and apoptosis by activating the PI3K/AKT signaling pathway. AMPK activation and NF-κB inhibition could inhibit AVICs calcification induced by PM treatment; however, AMPK and AKT inhibition reversed the protective effect of metformin. CONCLUSIONS This study, for the first time, demonstrates that metformin can inhibit AVICs in vitro calcification by activating the PI3K/AKT signaling pathway; this suggests that metformin may provide a potential target for the treatment of CAVD. And the PI3K/AKT signaling pathway emerges as an important regulatory axis in the pathological mechanism of CAVD.
Collapse
|
27
|
Bartoli-Leonard F, Zimmer J, Aikawa E. Innate and adaptive immunity: the understudied driving force of heart valve disease. Cardiovasc Res 2021; 117:2506-2524. [PMID: 34432007 PMCID: PMC8783388 DOI: 10.1093/cvr/cvab273] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Calcific aortic valve disease (CAVD), and its clinical manifestation that is calcific aortic valve stenosis, is the leading cause for valve disease within the developed world, with no current pharmacological treatment available to delay or halt its progression. Characterized by progressive fibrotic remodelling and subsequent pathogenic mineralization of the valve leaflets, valve disease affects 2.5% of the western population, thus highlighting the need for urgent intervention. Whilst the pathobiology of valve disease is complex, involving genetic factors, lipid infiltration, and oxidative damage, the immune system is now being accepted to play a crucial role in pathogenesis and disease continuation. No longer considered a passive degenerative disease, CAVD is understood to be an active inflammatory process, involving a multitude of pro-inflammatory mechanisms, with both the adaptive and the innate immune system underpinning these complex mechanisms. Within the valve, 15% of cells evolve from haemopoietic origin, and this number greatly expands following inflammation, as macrophages, T lymphocytes, B lymphocytes, and innate immune cells infiltrate the valve, promoting further inflammation. Whether chronic immune infiltration or pathogenic clonal expansion of immune cells within the valve or a combination of the two is responsible for disease progression, it is clear that greater understanding of the immune systems role in valve disease is required to inform future treatment strategies for control of CAVD development.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jonas Zimmer
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
28
|
Vidula MK, Orlenko A, Zhao L, Salvador L, Small AM, Horton E, Cohen JB, Adusumalli S, Denduluri S, Kobayashi T, Hyman M, Fiorilli P, Magro C, Singh B, Pourmussa B, Greczylo C, Basso M, Ebert C, Yarde M, Li Z, Cvijic ME, Wang Z, Walsh A, Maranville J, Kick E, Luettgen J, Adam L, Schafer P, Ramirez-Valle F, Seiffert D, Moore JH, Gordon D, Chirinos JA. Plasma biomarkers associated with adverse outcomes in patients with calcific aortic stenosis. Eur J Heart Fail 2021; 23:2021-2032. [PMID: 34632675 DOI: 10.1002/ejhf.2361] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 12/25/2022] Open
Abstract
AIMS Enhanced risk stratification of patients with aortic stenosis (AS) is necessary to identify patients at high risk for adverse outcomes, and may allow for better management of patient subgroups at high risk of myocardial damage. The objective of this study was to identify plasma biomarkers and multimarker profiles associated with adverse outcomes in AS. METHODS AND RESULTS We studied 708 patients with calcific AS and measured 49 biomarkers using a Luminex platform. We studied the correlation between biomarkers and the risk of (i) death and (ii) death or heart failure-related hospital admission (DHFA). We also utilized machine-learning methods (a tree-based pipeline optimizer platform) to develop multimarker models associated with the risk of death and DHFA. In this cohort with a median follow-up of 2.8 years, multiple biomarkers were significantly predictive of death in analyses adjusted for clinical confounders, including tumour necrosis factor (TNF)-α [hazard ratio (HR) 1.28, P < 0.0001], TNF receptor 1 (TNFRSF1A; HR 1.38, P < 0.0001), fibroblast growth factor (FGF)-23 (HR 1.22, P < 0.0001), N-terminal pro B-type natriuretic peptide (NT-proBNP) (HR 1.58, P < 0.0001), matrix metalloproteinase-7 (HR 1.24, P = 0.0002), syndecan-1 (HR 1.27, P = 0.0002), suppression of tumorigenicity-2 (ST2) (IL1RL1; HR 1.22, P = 0.0002), interleukin (IL)-8 (CXCL8; HR 1.22, P = 0.0005), pentraxin (PTX)-3 (HR 1.17, P = 0.001), neutrophil gelatinase-associated lipocalin (LCN2; HR 1.18, P < 0.0001), osteoprotegerin (OPG) (TNFRSF11B; HR 1.26, P = 0.0002), and endostatin (COL18A1; HR 1.28, P = 0.0012). Several biomarkers were also significantly predictive of DHFA in adjusted analyses including FGF-23 (HR 1.36, P < 0.0001), TNF-α (HR 1.26, P < 0.0001), TNFR1 (HR 1.34, P < 0.0001), angiopoietin-2 (HR 1.26, P < 0.0001), syndecan-1 (HR 1.23, P = 0.0006), ST2 (HR 1.27, P < 0.0001), IL-8 (HR 1.18, P = 0.0009), PTX-3 (HR 1.18, P = 0.0002), OPG (HR 1.20, P = 0.0013), and NT-proBNP (HR 1.63, P < 0.0001). Machine-learning multimarker models were strongly associated with adverse outcomes (mean 1-year probability of death of 0%, 2%, and 60%; mean 1-year probability of DHFA of 0%, 4%, 97%; P < 0.0001). In these models, IL-6 (a biomarker of inflammation) and FGF-23 (a biomarker of calcification) emerged as the biomarkers of highest importance. CONCLUSIONS Plasma biomarkers are strongly associated with the risk of adverse outcomes in patients with AS. Biomarkers of inflammation and calcification were most strongly related to prognosis.
Collapse
Affiliation(s)
- Mahesh K Vidula
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Alena Orlenko
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lei Zhao
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | - Lisa Salvador
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | - Aeron M Small
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Edward Horton
- Department of Internal Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jordana B Cohen
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Srinath Adusumalli
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Srinivas Denduluri
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Taisei Kobayashi
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Hyman
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Fiorilli
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Magro
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bibi Singh
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bianca Pourmussa
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Candy Greczylo
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael Basso
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | | | - Melissa Yarde
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | - Zhuyin Li
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | | | - Zhaoqing Wang
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | - Alice Walsh
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | | | - Ellen Kick
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | | | - Leonard Adam
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | - Peter Schafer
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | | | | | - Jason H Moore
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David Gordon
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | - Julio A Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
29
|
Engineering the aortic valve extracellular matrix through stages of development, aging, and disease. J Mol Cell Cardiol 2021; 161:1-8. [PMID: 34339757 DOI: 10.1016/j.yjmcc.2021.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 02/01/2023]
Abstract
For such a thin tissue, the aortic valve possesses an exquisitely complex, multi-layered extracellular matrix (ECM), and disruptions to this structure constitute one of the earliest hallmarks of fibrocalcific aortic valve disease (CAVD). The native valve structure provides a challenging target for engineers to mimic, but the development of advanced, ECM-based scaffolds may enable mechanistic and therapeutic discoveries that are not feasible in other culture or in vivo platforms. This review first discusses the ECM changes that occur during heart valve development, normal aging, onset of early-stage disease, and progression to late-stage disease. We then provide an overview of the bottom-up tissue engineering strategies that have been used to mimic the valvular ECM, and opportunities for advancement in these areas.
Collapse
|
30
|
Yang R, Tang Y, Chen X, Yang Y. Telocytes-derived extracellular vesicles alleviate aortic valve calcification by carrying miR-30b. ESC Heart Fail 2021; 8:3935-3946. [PMID: 34165260 PMCID: PMC8497371 DOI: 10.1002/ehf2.13460] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/13/2021] [Accepted: 05/23/2021] [Indexed: 12/28/2022] Open
Abstract
AIMS Calcific aortic valve disease (CAVD) is frequent in the elderly. Telocytes (TCs) are implicated in intercellular communication by releasing extracellular vesicles (EVs). This study investigated the role of TC-EVs in aortic valve calcification. METHODS AND RESULTS TCs were obtained and identified using enzymolysis method and flow cytometry. EVs were isolated from TCs using differential high-speed centrifugation method and identified using transmission electron microscope, western blot, and qNano analysis. The mouse model of CAVD was established. The changes of aortic valve activity-related indicators were analysed by ultrasound, and the expressions of TC markers CD34 and vimentin in mouse valve tissues were detected using RT-qPCR and western blot. The model mice were injected with TC-derived EVs. The expressions of Runx2, osteocalcin, and caspase-3 were detected using RT-qPCR and western blot. The calcification model of valvular interstitial cells (VICs) was established. TC-EVs were co-cultured with calcified VICs, and calcium deposition was detected using alizarin red S staining. miR-30b expression in calcified valvular tissues and cells was detected after EV treatment. miR-30b expression in TCs was knocked down and then EVs were extracted and co-cultured with calcified VICs. The target of miR-30b was predicted through bioinformatics website and verified using dual-luciferase assay. The levels of Wnt/β-catenin pathway-related proteins were detected. ApoE-/- mice fed with a high-fat diet showed decreased aortic valve orifice area, increased aortic transvalvular pressure difference and velocity, reduced left ventricular ejection fraction, decreased CD34 and vimentin, and increased caspase-3, Runx2, and osteocalcin. The levels of apoptosis- and osteogenesis- related proteins were inhibited after EV treatment. TC-EVs reduced calcium deposition and osteogenic proteins in calcified VICs. EVs could be absorbed by VICs. miR-30b expression was promoted in calcified valvular tissues and cells after EV treatment. Knockdown of miR-30b weakened the inhibitory effects of TC-EVs on calcium deposition and osteogenic proteins. miR-30b targeted Runx2. EV treatment inhibited the Wnt/β-catenin pathway, and knockdown of miR-30b in TCs attenuated the inhibitory effect of TC-EVs on the Wnt/β-catenin pathway. CONCLUSION TC-EVs played a protective role in aortic valve calcification via the miR-30b/Runx2/Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Rong Yang
- Department of Rheumatology, The Affiliated Zhongda Hospital, Southeast University, Nanjing, China
| | - Yihu Tang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Xiaowen Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Tandon I, Quinn KP, Balachandran K. Label-Free Multiphoton Microscopy for the Detection and Monitoring of Calcific Aortic Valve Disease. Front Cardiovasc Med 2021; 8:688513. [PMID: 34179147 PMCID: PMC8226007 DOI: 10.3389/fcvm.2021.688513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common valvular heart disease. CAVD results in a considerable socio-economic burden, especially considering the aging population in Europe and North America. The only treatment standard is surgical valve replacement as early diagnostic, mitigation, and drug strategies remain underdeveloped. Novel diagnostic techniques and biomarkers for early detection and monitoring of CAVD progression are thus a pressing need. Additionally, non-destructive tools are required for longitudinal in vitro and in vivo assessment of CAVD initiation and progression that can be translated into clinical practice in the future. Multiphoton microscopy (MPM) facilitates label-free and non-destructive imaging to obtain quantitative, optical biomarkers that have been shown to correlate with key events during CAVD progression. MPM can also be used to obtain spatiotemporal readouts of metabolic changes that occur in the cells. While cellular metabolism has been extensively explored for various cardiovascular disorders like atherosclerosis, hypertension, and heart failure, and has shown potential in elucidating key pathophysiological processes in heart valve diseases, it has yet to gain traction in the study of CAVD. Furthermore, MPM also provides structural, functional, and metabolic readouts that have the potential to correlate with key pathophysiological events in CAVD progression. This review outlines the applicability of MPM and its derived quantitative metrics for the detection and monitoring of early CAVD progression. The review will further focus on the MPM-detectable metabolic biomarkers that correlate with key biological events during valve pathogenesis and their potential role in assessing CAVD pathophysiology.
Collapse
Affiliation(s)
- Ishita Tandon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
32
|
LncRNA AFAP1-AS1 promotes M1 polarization of macrophages and osteogenic differentiation of valve interstitial cells. J Physiol Biochem 2021; 77:461-468. [PMID: 34043161 DOI: 10.1007/s13105-021-00821-0] [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: 12/16/2020] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
Little is known about the biological functions and underlying mechanisms of long non-coding RNA AFAP1-AS1 in degenerative calcified aortic valve disease (DCAVD). This study aims to explore whether AFAP1-AS1 regulates macrophage polarization in aortic valve calcification. Macrophage polarization and AFAP1-AS1 expression were detected in normal and calcified aortic valves of DCAVD patients. To explore the effect of AFAP1-AS1 on macrophage polarization, gain and loss of function were performed in THP-1 cells, and the percentage of M1 and M2 and the expressions of M1 and M2 markers were analyzed. Meanwhile, osteogenic differentiation was examined in valve interstitial cells (VICs). Compared with normal valves, there were more M1, less M2, and high AFAP1-AS1 expressions in calcified aortic valves, which may indicate a relationship between AFAP1-AS1 and macrophage polarization. AFAP1-AS1 overexpression promoted M1 polarization in lipopolysaccharide (LPS) and interferon gamma (IFN-γ)-treated THP-1 cells but inhibited M2 polarization, as well as augmented VIC osteogenic differentiation. On the contrary, the silence of AFAP1-AS1 could induce macrophage to M2-type and inhibit VIC osteogenic differentiation. These results elucidate that AFAP1-AS1 can promote M1 macrophages polarization to aggravate VIC osteogenic differentiation, playing a role in aortic valve calcification.
Collapse
|
33
|
Driscoll K, Cruz AD, Butcher JT. Inflammatory and Biomechanical Drivers of Endothelial-Interstitial Interactions in Calcific Aortic Valve Disease. Circ Res 2021; 128:1344-1370. [PMID: 33914601 DOI: 10.1161/circresaha.121.318011] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Calcific aortic valve disease is dramatically increasing in global burden, yet no therapy exists outside of prosthetic replacement. The increasing proportion of younger and more active patients mandates alternative therapies. Studies suggest a window of opportunity for biologically based diagnostics and therapeutics to alleviate or delay calcific aortic valve disease progression. Advancement, however, has been hampered by limited understanding of the complex mechanisms driving calcific aortic valve disease initiation and progression towards clinically relevant interventions.
Collapse
Affiliation(s)
| | - Alexander D Cruz
- Meinig School of Biomedical Engineering, Cornell University, Ithaca NY
| | | |
Collapse
|
34
|
Woodward HJ, Zhu D, Hadoke PWF, MacRae VE. Regulatory Role of Sex Hormones in Cardiovascular Calcification. Int J Mol Sci 2021; 22:4620. [PMID: 33924852 PMCID: PMC8125640 DOI: 10.3390/ijms22094620] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Sex differences in cardiovascular disease (CVD), including aortic stenosis, atherosclerosis and cardiovascular calcification, are well documented. High levels of testosterone, the primary male sex hormone, are associated with increased risk of cardiovascular calcification, whilst estrogen, the primary female sex hormone, is considered cardioprotective. Current understanding of sexual dimorphism in cardiovascular calcification is still very limited. This review assesses the evidence that the actions of sex hormones influence the development of cardiovascular calcification. We address the current question of whether sex hormones could play a role in the sexual dimorphism seen in cardiovascular calcification, by discussing potential mechanisms of actions of sex hormones and evidence in pre-clinical research. More advanced investigations and understanding of sex hormones in calcification could provide a better translational outcome for those suffering with cardiovascular calcification.
Collapse
Affiliation(s)
- Holly J. Woodward
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK;
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Patrick W. F. Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK;
| | - Victoria E. MacRae
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK;
| |
Collapse
|
35
|
Hu W, Wu R, Gao C, Liu F, Zeng Z, Zhu Q, Chen J, Cheng S, Yu K, Qian Y, Zhao J, Zhong S, Li Q, Wang L, Liu X, Wang J. Knockdown of estrogen-related receptor α inhibits valve interstitial cell calcification in vitro by regulating heme oxygenase 1. FASEB J 2021; 35:e21183. [PMID: 33184978 DOI: 10.1096/fj.202001588rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/15/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022]
Abstract
Calcific aortic valve disease (CAVD) is the most common valvular heart disease in adults. The cellular mechanisms of CAVD are still unknown, but accumulating evidence has revealed that osteogenic differentiation of human valve interstitial cells (hVICs) plays an important role in CAVD. Thus, we aimed to investigate the function of estrogen-related receptor α (ERRα) in the osteogenic differentiation of hVICs. We found that the level of ERRα was significantly increased in CAVD samples compared to normal controls. In addition, ERRα was significantly upregulated during hVIC osteogenic differentiation in vitro. Gain- and loss-of-function experiments were performed to identify the function of ERRα in hVIC calcification in vitro. Inhibition of endogenous ERRα attenuated hVIC calcification, whereas overexpression of ERRα in hVICs promoted this process. RNA sequencing results suggested that heme oxygenase-1 (Hmox1) was a downstream target of ERRα, which was further confirmed by western blotting. Additionally, we also found that downregulation of Hmox1 by shHmox1 efficiently reversed the inhibition of calcification induced by ERRα shRNA in hVICs. ChIP-qPCR and luciferase assays indicated that Hmox1 was negatively regulated by ERRα. We found that overexpression of Hmox1 or its substrates significantly inhibited hVIC calcification in vitro. In conclusion, we found that knockdown of ERRα can inhibit hVIC calcification through upregulating Hmox1 and that ERRα and Hmox1 are potential targets for the treatment of CAVD.
Collapse
Affiliation(s)
- Wangxing Hu
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Rongrong Wu
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Chenyang Gao
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Feng Liu
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Zhiru Zeng
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Qifeng Zhu
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Jinyong Chen
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Si Cheng
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Kaixiang Yu
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Yi Qian
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Jing Zhao
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Shuhan Zhong
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Qingju Li
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Lihan Wang
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Xianbao Liu
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| | - Jian'an Wang
- Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cardiovascular Disease of Zhejiang Province, Hangzhou, China
| |
Collapse
|
36
|
Tandon I, Johns S, Woessner A, Perez J, Cross D, Ozkizilcik A, Muldoon TJ, Vallurupalli S, Padala M, Quinn KP, Balachandran K. Label-free optical biomarkers detect early calcific aortic valve disease in a wild-type mouse model. BMC Cardiovasc Disord 2020; 20:521. [PMID: 33308143 PMCID: PMC7731510 DOI: 10.1186/s12872-020-01776-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/08/2020] [Indexed: 12/31/2022] Open
Abstract
Background Calcific aortic valve disease (CAVD) pathophysiology is a complex, multistage process, usually diagnosed at advanced stages after significant anatomical and hemodynamic changes in the valve. Early detection of disease progression is thus pivotal in the development of prevention and mitigation strategies. In this study, we developed a diet-based, non-genetically modified mouse model for early CAVD progression, and explored the utility of two-photon excited fluorescence (TPEF) microscopy for early detection of CAVD progression. TPEF imaging provides label-free, non-invasive, quantitative metrics with the potential to correlate with multiple stages of CAVD pathophysiology including calcium deposition, collagen remodeling and osteogenic differentiation. Methods Twenty-week old C57BL/6J mice were fed either a control or pro-calcific diet for 16 weeks and monitored via echocardiography, histology, immunohistochemistry, and quantitative polarized light imaging. Additionally, TPEF imaging was used to quantify tissue autofluorescence (A) at 755 nm, 810 nm and 860 nm excitation, to calculate TPEF 755–860 ratio (A860/525/(A755/460 + A860/525)) and TPEF Collagen-Calcium ratio (A810/525/(A810/460 + A810/525)) in the murine valves. In a separate experiment, animals were fed the above diets till 28 weeks to assess for later-stage calcification. Results Pro-calcific mice showed evidence of lipid deposition at 4 weeks and calcification at 16 weeks at the valve commissures. The valves of pro-calcific mice also showed positive expression for markers of osteogenic differentiation, myofibroblast activation, proliferation, inflammatory cytokines and collagen remodeling. Pro-calcific mice exhibited lower TPEF autofluorescence ratios, at locations coincident with calcification, that correlated with increased collagen disorganization and positive expression of osteogenic markers. Additionally, locations with lower TPEF autofluorescence ratios at 4 and 16 weeks exhibited increased calcification at later 28-week timepoints. Conclusions This study suggests the potential of TPEF autofluorescence metrics to serve as a label-free tool for early detection and monitoring of CAVD pathophysiology.
Collapse
Affiliation(s)
- Ishita Tandon
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Shelby Johns
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Alan Woessner
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Jessica Perez
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Delaney Cross
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Asya Ozkizilcik
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Timothy J Muldoon
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Srikanth Vallurupalli
- Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Muralidhar Padala
- Division of Cardiothoracic Surgery, Joseph P. Whitehead Department of Surgery, Emory University, Atlanta, GA, 30322, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA
| | - Kartik Balachandran
- Department of Biomedical Engineering, University of Arkansas, 122 John A. White Jr. Engineering Hall, Fayetteville, AR, 72701, USA.
| |
Collapse
|
37
|
Abstract
Familial hypercholesterolemia (FH) is a rare autosomal gene deficiency disease with increased low-density lipoprotein cholesterol, xanthoma, and premature coronary heart disease. Calcified aortic valve disease (CAVD) is prevalent in FH patients, resulting in adverse events and heavy health care burden. Aortic valve calcification is currently considered an active biological process, which shares several common risk factors with atherosclerosis, including aging, hypertension, dyslipidemia, and so on. Unfortunately, the pathogenesis and therapy of CAVD in FH are still controversial. There is no pharmacological intervention recommended to delay the development of CAVD in FH, and the only effective treatment for severe CAVD is aortic valve replacement. In this review, we summarize the detailed description of the pathophysiology, molecular mechanism, risk factors, and treatment of CAVD in FH patients.
Collapse
|
38
|
AGEs-RAGE axis causes endothelial-to-mesenchymal transition in early calcific aortic valve disease via TGF-β1 and BMPR2 signaling. Exp Gerontol 2020; 141:111088. [DOI: 10.1016/j.exger.2020.111088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/24/2020] [Accepted: 09/03/2020] [Indexed: 01/08/2023]
|
39
|
miR-214 Attenuates Aortic Valve Calcification by Regulating Osteogenic Differentiation of Valvular Interstitial Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:971-980. [PMID: 33251046 PMCID: PMC7679242 DOI: 10.1016/j.omtn.2020.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/10/2020] [Indexed: 01/04/2023]
Abstract
Calcific aortic valve disease (CAVD) is a common heart valve disease in aging populations, and aberrant osteogenic differentiation of valvular interstitial cells (VICs) plays a critical role in the pathogenesis of ectopic ossification of the aortic valve. miR-214 has been validated to be involved in the osteogenesis process. Here, we aim to investigate the role and mechanism of miR-214 in CAVD progression. miR-214 expression was significantly downregulated in CAVD aortic valve leaflets, accompanied by upregulation of osteogenic markers. Overexpression of miR-214 suppressed osteogenic differentiation of VICs, while silencing the expression of miR-214 promoted this function. miR-214 directly targeted ATF4 and Sp7 to modulate osteoblastic differentiation of VICs, which was proved by dual luciferase reporter assay and rescue experiment. miR-214 knockout rats exhibited higher mean transvalvular velocity and gradient. The expression of osteogenic markers in aortic valve leaflets of miR-214 knockout rats was upregulated compared to that of the wild-type group. Taken together, our study showed that miR-214 inhibited aortic valve calcification via regulating osteogenic differentiation of VICs by directly targeting ATF4 and Sp7, indicating that miR-214 may act as a profound candidate of targeting therapy for CAVD.
Collapse
|
40
|
MacGrogan D, Martínez-Poveda B, Desvignes JP, Fernandez-Friera L, Gomez MJ, Gil Vilariño E, Callejas Alejano S, Garcia-Pavia P, Solis J, Lucena J, Salgado D, Collod-Béroud G, Faure E, Théron A, Torrents J, Avierinos JF, Montes L, Dopazo A, Fuster V, Ibañez B, Sánchez-Cabo F, Zaffran S, de la Pompa JL. Identification of a peripheral blood gene signature predicting aortic valve calcification. Physiol Genomics 2020; 52:563-574. [PMID: 33044885 DOI: 10.1152/physiolgenomics.00034.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is a significant cause of illness and death worldwide. Identification of early predictive markers could help optimize patient management. RNA-sequencing was carried out on human fetal aortic valves at gestational weeks 9, 13, and 22 and on a case-control study with adult noncalcified and calcified bicuspid and tricuspid aortic valves. In dimension reduction and clustering analyses, diseased valves tended to cluster with fetal valves at week 9 rather than normal adult valves, suggesting that part of the disease program might be due to reiterated developmental processes. The analysis of groups of coregulated genes revealed predominant immune-metabolic signatures, including innate and adaptive immune responses involving lymphocyte T-cell metabolic adaptation. Cytokine and chemokine signaling, cell migration, and proliferation were all increased in CAVD, whereas oxidative phosphorylation and protein translation were decreased. Discrete immune-metabolic gene signatures were present at fetal stages and increased in adult controls, suggesting that these processes intensify throughout life and heighten in disease. Cellular stress response and neurodegeneration gene signatures were aberrantly expressed in CAVD, pointing to a mechanistic link between chronic inflammation and biological aging. Comparison of the valve RNA-sequencing data set with a case-control study of whole blood transcriptomes from asymptomatic individuals with early aortic valve calcification identified a highly predictive gene signature of CAVD and of moderate aortic valve calcification in overtly healthy individuals. These data deepen and broaden our understanding of the molecular basis of CAVD and identify a peripheral blood gene signature for the early detection of aortic valve calcification.
Collapse
Affiliation(s)
- Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Martínez-Poveda
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Jean-Pierre Desvignes
- Aix Marseille University, Marseille Medical Genetics, INSERM U1251, Marseille, France
| | - Leticia Fernandez-Friera
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain.,Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,HM Hospitales-Centro Integral de Enfermedades Cardiovasculares, Madrid, Spain
| | - Manuel José Gomez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Eduardo Gil Vilariño
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Sergio Callejas Alejano
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Pablo Garcia-Pavia
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain.,Departmento de Cardiología, Hospital Universitario Puerta de Hierro, Madrid, Spain.,Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Jorge Solis
- Departmento of Cardiología, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Joaquín Lucena
- Servicio de Patología Forense, Instituto de Medicina Legal y Ciencias Forenses
| | - David Salgado
- Aix Marseille University, Marseille Medical Genetics, INSERM U1251, Marseille, France
| | | | - Emilie Faure
- Aix Marseille University, Marseille Medical Genetics, INSERM U1251, Marseille, France
| | - Alexis Théron
- Service de Cardiologie, Hôpital de la Timone, Marseille, France
| | - Julia Torrents
- Service d'anatomie et Cytologie Pathologiques, Hôpital de la Timone, Marseille, France
| | - Jean-François Avierinos
- Aix Marseille University, Marseille Medical Genetics, INSERM U1251, Marseille, France.,Service de Cardiologie, Hôpital de la Timone, Marseille, France
| | | | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Valentín Fuster
- Cardiovascular Imaging and Population Studies Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Cardiology Department, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Borja Ibañez
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain.,Service d'anatomie et Cytologie Pathologiques, Hôpital de la Timone, Marseille, France.,Hospital Clínico San Carlos, Madrid, Spain.,IIS-Fundación Jiménez Díaz Hospital Universitario, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Stephane Zaffran
- Aix Marseille University, Marseille Medical Genetics, INSERM U1251, Marseille, France
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
41
|
Patient Management in Aortic Stenosis: Towards Precision Medicine Through Protein Analysis, Imaging and Diagnostic Tests. J Clin Med 2020; 9:jcm9082421. [PMID: 32731585 PMCID: PMC7463596 DOI: 10.3390/jcm9082421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 01/12/2023] Open
Abstract
Aortic stenosis is the most frequent valvular disease in developed countries. It progresses from mild fibrocalcific leaflet changes to a more severe leaflet calcification at the end stages of the disease. Unfortunately, symptoms of aortic stenosis are unspecific and only appear when it is too late, complicating patients' management. The global impact of aortic stenosis is increasing due to the growing elderly population. The disease supposes a great challenge because of the multiple comorbidities of these patients. Nowadays, the only effective treatment is valve replacement, which has a high cost in both social and economic terms. For that reason, it is crucial to find potential diagnostic, prognostic and therapeutic indicators that could help us to detect this disease in its earliest stages. In this article, we comprehensively review several key observations and translational studies related to protein markers that are promising for being implemented in the clinical field as well as a discussion about the role of precision medicine in aortic stenosis.
Collapse
|
42
|
Strianese O, Rizzo F, Ciccarelli M, Galasso G, D’Agostino Y, Salvati A, Del Giudice C, Tesorio P, Rusciano MR. Precision and Personalized Medicine: How Genomic Approach Improves the Management of Cardiovascular and Neurodegenerative Disease. Genes (Basel) 2020; 11:E747. [PMID: 32640513 PMCID: PMC7397223 DOI: 10.3390/genes11070747] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Life expectancy has gradually grown over the last century. This has deeply affected healthcare costs, since the growth of an aging population is correlated to the increasing burden of chronic diseases. This represents the interesting challenge of how to manage patients with chronic diseases in order to improve health care budgets. Effective primary prevention could represent a promising route. To this end, precision, together with personalized medicine, are useful instruments in order to investigate pathological processes before the appearance of clinical symptoms and to guide physicians to choose a targeted therapy to manage the patient. Cardiovascular and neurodegenerative diseases represent suitable models for taking full advantage of precision medicine technologies applied to all stages of disease development. The availability of high technology incorporating artificial intelligence and advancement progress made in the field of biomedical research have been substantial to understand how genes, epigenetic modifications, aging, nutrition, drugs, microbiome and other environmental factors can impact health and chronic disorders. The aim of the present review is to address how precision and personalized medicine can bring greater clarity to the clinical and biological complexity of these types of disorders associated with high mortality, involving tremendous health care costs, by describing in detail the methods that can be applied. This might offer precious tools for preventive strategies and possible clues on the evolution of the disease and could help in predicting morbidity, mortality and detecting chronic disease indicators much earlier in the disease course. This, of course, will have a major effect on both improving the quality of care and quality of life of the patients and reducing time efforts and healthcare costs.
Collapse
Affiliation(s)
- Oriana Strianese
- Clinical Research and Innovation, Clinica Montevergine S.p.A., 83013 Mercogliano, Italy; (O.S.); (C.D.G.)
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (F.R.); (Y.D.); (A.S.)
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (F.R.); (Y.D.); (A.S.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (M.C.); (G.G.)
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (M.C.); (G.G.)
| | - Ylenia D’Agostino
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (F.R.); (Y.D.); (A.S.)
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (F.R.); (Y.D.); (A.S.)
| | - Carmine Del Giudice
- Clinical Research and Innovation, Clinica Montevergine S.p.A., 83013 Mercogliano, Italy; (O.S.); (C.D.G.)
| | - Paola Tesorio
- Unit of Cardiology, Clinica Montevergine S.p.A., 83013 Mercogliano, Italy;
| | - Maria Rosaria Rusciano
- Clinical Research and Innovation, Clinica Montevergine S.p.A., 83013 Mercogliano, Italy; (O.S.); (C.D.G.)
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84084 Baronissi, Italy; (M.C.); (G.G.)
| |
Collapse
|
43
|
Association of aortic valve calcification with carotid artery lesions and peripheral artery disease in patients with chronic kidney disease: a cross-sectional study. BMC Nephrol 2020; 21:203. [PMID: 32471374 PMCID: PMC7260754 DOI: 10.1186/s12882-020-01864-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/21/2020] [Indexed: 11/10/2022] Open
Abstract
Background Patients with chronic kidney disease (CKD) reportedly have a high prevalence of aortic valve calcification (AVC). In population-based studies, AVC is considered a manifestation of systemic atherosclerosis. The association of AVC with atherosclerotic lesions has not been fully investigated in predialysis patients. The present study was performed to determine whether carotid artery lesions and peripheral artery disease (PAD) are associated with AVC in patients with CKD not on dialysis. Methods In total, 749 patients were included in this cross-sectional study. AVC was evaluated using echocardiography. Carotid artery lesions including carotid artery plaque (CAP) and PAD were simultaneously examined in each patient. A logistic regression analysis was applied to determine the factors associated with AVC. Results AVC, CAP, and PAD were found in 201, 583, and 123 patients, respectively. In the multivariable analyses adjusted for covariates including the estimated glomerular filtration rate and makers of mineral metabolism (serum calcium, serum phosphorus, parathyroid hormone, 1,25-dihydroxyvitamin D, and fibroblast growth factor 23), AVC was significantly associated with the presence of CAP [odds ratio (OR), 3.37; 95% confidence interval (CI), 1.43–7.95], the presence of PAD (OR, 1.76; 95% CI, 1.10–2.81), the CAP score (per 1.0-point increase) (OR, 1.06; 95% CI, 1.02–1.11), and the ankle-brachial blood pressure index (per 0.1-point increase) (OR, 0.83; 95% CI, 0.72–0.95). Conclusions AVC was associated with atherosclerotic lesions independent of kidney function and mineral metabolism. We consider that this association between AVC and atherosclerosis might reflect the burden of shared atherosclerotic risk factors.
Collapse
|
44
|
Late Cerebrovascular Events Following Transcatheter Aortic Valve Replacement. JACC Cardiovasc Interv 2020; 13:872-881. [DOI: 10.1016/j.jcin.2019.11.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 11/23/2022]
|
45
|
Haberman D, Chernin G, Meledin V, Zikry M, Shuvy M, Gandelman G, Goland S, George J, Shimoni S. Urea level is an independent predictor of mortality in patients with severe aortic valve stenosis. PLoS One 2020; 15:e0230002. [PMID: 32160250 PMCID: PMC7065786 DOI: 10.1371/journal.pone.0230002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 02/19/2020] [Indexed: 11/19/2022] Open
Abstract
Introduction Severe aortic stenosis (AS) is the most common valvular heart disease in the western world. Various factors are related to severe AS prognosis, including chronic kidney disease. The aim of this study was to evaluate the prognostic value of urea level in patients with severe AS. Methods We prospectively enrolled 142 patients (79.1±9.4 years, 88 women) with severe AS (mean valve area 0.67± 0.17 cm2). Clinical assessment, blood tests and echocardiography were performed at enrollment and follow up. The patient population was divided into low and high urea level groups, according to the median urea level at enrollment (72 patients, mean urea 35.5±6.2 mg/dL and 70 patients, mean urea 61.1±17.8 mg/dL, respectively). Hundred and twelve patients (79%) underwent aortic valve intervention. The primary endpoint was all-cause and cardiovascular mortality. Outcomes During follow-up of 37±19.5 months, 56 (37.1%) patients died, 39 due to cardiovascular causes. In univariate analysis, age, urea level, creatinine, New York Heart Association (NYHA) class and aortic valve intervention were associated with all-cause mortality. However, in multivariate analysis only aortic valve intervention and blood urea were independent predictors of all-cause mortality (HR 0.494; 95% CI 0.226–0.918, P = 0.026 and HR 1.015; 95% CI 1.003–1.029, P = 0.046 respectively). Urea level, NYHA class and age were also significant predictors of cardiovascular mortality. Whereas, in multivariate analysis, only urea level predicted cardiovascular mortality in these patients (HR 1.017; CI 1.003–1.031 P = 0.019). Conclusions Blood urea, a generally readily available and routinely determined marker of renal function, is an independent prognostic factor in patients with severe AS.
Collapse
Affiliation(s)
- Dan Haberman
- The Heart Institute, Kaplan Medical Center, Rehovot, Israel
- Hebrew University and Hadassah Medical School, Jerusalem, Israel
- * E-mail:
| | - Gil Chernin
- Nephrology Department Kaplan Medical Center, Rehovot, Israel
- Heart Institute, Hadassah Medical Center, Jerusalem, Israel
| | - Valery Meledin
- The Heart Institute, Kaplan Medical Center, Rehovot, Israel
- Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Meital Zikry
- The Heart Institute, Kaplan Medical Center, Rehovot, Israel
- Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Mony Shuvy
- Hebrew University and Hadassah Medical School, Jerusalem, Israel
- Heart Institute, Hadassah Medical Center, Jerusalem, Israel
| | - Gera Gandelman
- The Heart Institute, Kaplan Medical Center, Rehovot, Israel
- Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Sorel Goland
- The Heart Institute, Kaplan Medical Center, Rehovot, Israel
- Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Jacob George
- The Heart Institute, Kaplan Medical Center, Rehovot, Israel
- Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Sara Shimoni
- The Heart Institute, Kaplan Medical Center, Rehovot, Israel
- Hebrew University and Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
46
|
Small AM, Kiss DH, Anwaruddin S, Giri J, Han Y, Zhao L, Salvador L, Cvijic ME, Li Z, Chirinos JA, Damrauer SM, Rader DJ. Soluble FMS-Like Tyrosine Kinase-1 Is a Circulating Biomarker Associated With Calcific Aortic Stenosis. J Am Coll Cardiol 2020; 73:1364-1365. [PMID: 30898214 DOI: 10.1016/j.jacc.2019.01.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/22/2018] [Accepted: 01/08/2019] [Indexed: 10/27/2022]
|
47
|
Hayashi H, Akiyama K, Itatani K, DeRoo S, Sanchez J, Ferrari G, Colombo PC, Takeda K, Wu IY, Kainuma A, Takayama H. A novel in vivo assessment of fluid dynamics on aortic valve leaflet using epi-aortic echocardiogram. Echocardiography 2020; 37:323-330. [PMID: 32003907 DOI: 10.1111/echo.14596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/27/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mechanical stress caused by blood flow, such as wall shear stress (WSS) and its related parameters, is key moderator of endothelial degeneration. However, an in vivo method to measure WSS on heart valves has not been developed. METHODS We developed a novel approach, based on vector flow mapping using intraoperative epi-aortic echocardiogram, to measure WSS and oscillatory shear index (OSI) on the aortic valve. We prospectively enrolled 15 patients with normal valves, who underwent coronary artery bypass graft. RESULTS Systolic WSS on the ventricularis (2.40 ± 0.44 Pa [1.45-3.00 Pa]) was higher than systolic WSS on the fibrosa (0.33 ± 0.08 Pa [0.14-0.47 Pa], P < .001) and diastolic WSS on the ventricularis (0.18 ± 0.07 Pa [0.04-0.28 Pa], P < .001). Oscillatory shear index on the fibrosa was higher than on the ventricularis (0.29 ± 0.04 [0.24-0.36] vs 0.05 ± 0.03 [0.01-0.12], P < .001). A pilot study involving two patients with severe aortic regurgitation showed significantly different values in fluid dynamics. CONCLUSION Vector flow mapping method using intraoperative epi-aortic echocardiogram is an effective way of measuring WSS and OSI on normal aortic leaflet in vivo, allowing for better understanding of the pathophysiology of aortic valve diseases.
Collapse
Affiliation(s)
- Hideyuki Hayashi
- Division of Cardiothoracic Surgery, Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - Koichi Akiyama
- Department of Anesthesiology, Yodogawa Christian Hospital, Osaka, Japan
| | - Keiichi Itatani
- Department of Cardiovascular Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Scott DeRoo
- Division of Cardiothoracic Surgery, Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - Joseph Sanchez
- Division of Cardiothoracic Surgery, Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - Giovanni Ferrari
- Department of Surgery and Biomedical Engineering, Columbia University Medical, New York, NY, USA
| | - Paolo C Colombo
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Koji Takeda
- Division of Cardiothoracic Surgery, Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - Isaac Y Wu
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Atsushi Kainuma
- Division of Cardiothoracic Surgery, Department of Surgery, Columbia University Medical Center, New York, NY, USA
| | - Hiroo Takayama
- Division of Cardiothoracic Surgery, Department of Surgery, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
48
|
Song R, Zhai Y, Ao L, Fullerton DA, Meng X. MicroRNA-204 Deficiency in Human Aortic Valves Elevates Valvular Osteogenic Activity. Int J Mol Sci 2019; 21:ijms21010076. [PMID: 31861929 PMCID: PMC6981435 DOI: 10.3390/ijms21010076] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Aortic valve interstitial cells (AVICs) play a major role in valvular calcification associated with calcific aortic valve disease (CAVD). Although AVICs from diseased valves display a pro-osteogenic phenotype, the underlying mechanism causing this remains unclear. MicroRNA-204 (miR-204) is a negative regulator of osteoblast differentiation. We sought to analyze miR-204 expression in diseased human aortic valves and determine the role of this miR in AVIC osteogenic activity associated with CAVD pathobiology. In situ hybridization and PCR analysis revealed miR-204 deficiency in diseased valves and in AVICs from diseased valves. MiR-204 mimic suppressed alkaline phosphatase (ALP) expression and calcium deposition in AVICs from diseased valves. MiR-204 antagomir enhanced ALP expression in AVICs from normal valves through induction of Runx2 and Osx, and expression of miR-204 antagomir in mouse aortic valves promoted calcium deposition through up-regulation of Runx2 and Osx. Further, miR-204 mimic suppressed the osteogenic responses to TGF-β1 in AVICs of normal valves. In conclusion, miR-204 deficiency contributes to the mechanism underlying elevated osteogenic activity in diseased aortic valves, and miR-204 is capable of reversing the pro-osteogenic phenotype of AVICs of diseased valves and suppressing AVIC osteogenic response to stimulation. Exogenous miR-204 may have therapeutic potential for inhibiting valvular calcification associated with CAVD progression.
Collapse
|
49
|
Khan K, Albanese I, Yu B, Shalal Y, Al-Kindi H, Alaws H, Tardif JC, Gourgas O, Cerutti M, Schwertani A. Urotensin II, urotensin-related peptide, and their receptor in aortic valve stenosis. J Thorac Cardiovasc Surg 2019; 161:e1-e15. [PMID: 31679703 DOI: 10.1016/j.jtcvs.2019.09.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Aortic valve stenosis (AVS) is the most common cause of surgical valve replacement worldwide. The vasoactive peptide urotensin II (UII) is upregulated in atherosclerosis and several other cardiovascular diseases; however, its role in the pathogenesis of AVS remains to be determined. Here, we investigated the expression of UII, urotensin-related peptide (URP), and the urotensin receptor (UT) and the role this system plays in AVS. METHODS Immunohistochemistry and reverse-transcriptase polymerase chain reaction were used to examine the cellular localization and mRNA expression, of UII, URP, and UT in calcified and noncalcified aortic valves. Human aortic valve interstitial cells were isolated from normal valves and treated with UII or URP, and changes in cell proliferation, cholesterol efflux, calcium deposition, and β-catenin translocation were assessed. RESULTS The mRNA expression of UII, URP, and UT was significantly greater in patients with AVS. There was abundant presence of UII, URP, and UT immunostaining in diseased compared with nondiseased valves and correlated significantly with presence of calcification (P < .0001) and fibrosis (P < .0001). Treating human aortic valve interstitial cells with UII or URP significantly increased cell proliferation (P < .0001) and decreased cholesterol efflux (P = .0011 and P = .0002, respectively). UII also significantly reduced ABCA1 protein expression (P = .0457) and increased β-catenin nuclear translocation (P < .0001) and mineral deposition (P < .0001). CONCLUSIONS Together, these data suggest that the urotensin system plays a role in the pathogenesis of AVS and warrants further investigation.
Collapse
Affiliation(s)
- Kashif Khan
- Cardiology and Cardiac Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | - Isabella Albanese
- Cardiology and Cardiac Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | - Bin Yu
- Cardiology and Cardiac Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | - Yousif Shalal
- Cardiology and Cardiac Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | - Hamood Al-Kindi
- Cardiology and Cardiac Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | - Hossney Alaws
- Cardiology and Cardiac Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | | | - Ophélie Gourgas
- Department of Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - Marta Cerutti
- Department of Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - Adel Schwertani
- Cardiology and Cardiac Surgery, McGill University Health Center, Montreal, Quebec, Canada.
| |
Collapse
|
50
|
Affiliation(s)
- Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behavior (A.V.G.).,Department of Physiology and Membrane Biology (A.V.G.), University of California, Davis
| |
Collapse
|