1
|
Wang Y, Nie Z, Liu H, de Bruijn JD, Yuan H, Bao C. Apolipoprotein E as a Potential Regulator of Osteoclast-Osteoblast Coupling in Material-Induced Bone Formation. Acta Biomater 2025:S1742-7061(25)00405-2. [PMID: 40449705 DOI: 10.1016/j.actbio.2025.05.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 05/21/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Osteoinductive materials which induce bone formation in non-osseous sites are promising bone substitutes to repair critical-sized bone defects. It appears that innate immune response (esp. osteoclastogenesis) to materials plays an important role in material-induced bone formation. In this study, the coupling between osteoclastogenesis and subsequent osteogenesis in material-induced bone formation was investigated. Osteoclastogenesis of mouse bone marrow-derived monocytes (BMMs) on osteoinductive tricalcium phosphate (TCPs) and non-osteoinductive tricalcium phosphate (TCPb) ceramics were evaluated with high-throughput RNA sequencing (RNA-seq) and RT-qPCR regarding secretory proteins. It turned out that osteoinductive TCPs supported osteoclastogenesis and enhanced Apolipoprotein E (ApoE) production. Meanwhile, ApoE enhanced osteogenic gene expression (Alp, Runx2, Col1a1, Osterix) and ALP staining and activity of CRL-12424 cells in vitro. Additionally, western blot assay revealed that ApoE played its role in osteogenesis of CRL-12424 by activating JAK-STAT pathway instead of PI3K-AKT pathway. The overall data indicated that ApoE was a potential coupling factor between osteoclastogenesis and osteogenesis in material-induced bone formation. By secreting ApoE, osteoclasts formed on osteoinductive materials stimulated osteogenic differentiation of osteo-progenitors via JAK-STAT pathway. STATEMENT OF SIGNIFICANCE: Osteoinductive materials can repair critical-sized bone defects, while the precise mechanism osteoinductive materials driving bone formation remains unclear. Recent research has highlighted the role of osteoclastogenesis in material-induced bone formation, how osteoclastogenesis playing its role in osteogenesis was subjected to investigation in the current study. Robust ApoE gene expression shown in osteoclastogenesis with the osteoinductive material and ApoE enhancing osteogenesis of mesenchymal stromal cells (CRL-12424) indicated ApoE as a potential regulator of osteoclast-osteoblast coupling, providing thus novel insights into the complex interplay of cellular responses and contributing to the development of more effective bone substitute materials.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhangling Nie
- Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
| | - Huaze Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Joost D de Bruijn
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, 3723 MB Bilthoven, the Netherlands
| | - Huipin Yuan
- Kuros Biosciences BV, Prof. Bronkhorstlaan 10, 3723 MB Bilthoven, the Netherlands; Huipin Yuan's Lab, Sichuan, China.
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
2
|
Lai JC, Chang GRL, Tu MY, Cidem A, Chen IC, Chen CM. Potential of Kefir-Derived Peptides, Probiotics, and Exopolysaccharides for Osteoporosis Management. Curr Osteoporos Rep 2025; 23:18. [PMID: 40192921 PMCID: PMC11976759 DOI: 10.1007/s11914-025-00910-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/10/2025]
Abstract
PURPOSE OF REVIEW Osteoporosis is a prevalent skeletal disorder in postmenopausal women and older adults. Kefir has gained attention for its potent antioxidative, anti-inflammatory, and immunomodulatory properties. This review consolidates findings on kefir-derived peptides' interventions in osteoporosis models and evaluates the therapeutic potential of kefir components in preventing osteoporosis, thereby enhancing its application in clinical nutrition strategies for osteoporosis management. RECENT FINDINGS Kefir-derived peptides exhibit osteoprotective potential in various animal models of osteoporosis, in which several antioxidative and ACE-inhibitory peptides have been shown to promote osteoblast differentiation and mineralization. In addition, emerging evidence supports the role of kefir-derived probiotics and exopolysaccharides (kefiran) in mitigating bone loss. Kefir holds significant promise in the management of osteoporosis due to its unique composition of bioactive components promoting bone health. While research is still in its early stages, evidence suggests kefir's potential as a natural approach to osteoporosis prevention and management.
Collapse
Affiliation(s)
- Jen-Chieh Lai
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan
- Department of Orthopedic Surgery, Taichung Armed Forces General Hospital, Taichung, 411, Taiwan
| | - Gary Ro-Lin Chang
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan
| | - Min-Yu Tu
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan
- Department of Orthopedic Surgery, Taichung Armed Forces General Hospital, Taichung, 411, Taiwan
| | - Abdulkadir Cidem
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - I-Chien Chen
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
- The iEGG and Animal Biotechnology Research Center, National Chung Hsing University, Taichung, 402, Taiwan.
- Rong Hsing Research Center for Translational Medicine, Taichung Veterans General Hospital, Taichung, 407, Taiwan.
- Center for General Educational, National Quemoy University, Kinmen, 892, Taiwan.
| |
Collapse
|
3
|
Zhang J, Lei W, Zhou J, Zhang Y, Huang F, Chen M. Uric acid promotes aortic valve calcification via mediating valve interstitial cell osteogenic differentiation and endothelial dysfunction. FASEB J 2025; 39:e70437. [PMID: 40100089 DOI: 10.1096/fj.202402831r] [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: 12/04/2024] [Revised: 01/25/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Aortic valve calcification is a lethal valvular heart disease lacking effective drug therapy. However, whether uric acid is involved in the development of aortic valve calcification is unclear. Two-sample Mendelian randomization (MR) analyses confirmed the causal relationship between uric acid and valvular heart disease. Uric acid levels were assessed in aortic valve tissue from patients with/without aortic valve calcification. To investigate the impact of hyperuricemia on aortic valve calcification, apolipoprotein E knockout (ApoE-/-) mice fed a high-fat diet (HFD) were also given an adenine diet, with some receiving allopurinol in their drinking water. RNA sequencing was performed on valve interstitial cells (VICs) and endothelial cells (VECs) with/without uric acid. MR analysis has revealed a causal effect of uric acid levels on valvular heart disease. Furthermore, our clinical data indicate a positive correlation between elevated serum uric acid levels and aortic valve calcium score. Specifically, uric acid levels were upregulated in calcified valves. In ApoE-/- mice, an adenine-diet-induced hyperuricemia accelerated aortic valve calcification. RNA sequencing analysis demonstrated that uric acid-promoted osteogenic differentiation, primarily through the activation of hypoxia-inducible factor-1alpha (HIF-α). Additionally, uric acid impaired endothelial barrier function by activating HIF-α, resulting in increased macrophage infiltration in ApoE-/- mice. Inhibiting HIF-1α suppressed osteogenic differentiation and reduced endothelial injury both in vitro and in vivo in the presence of uric acid. This study reveals a new role of hyperuricemia in aortic valve calcification, suggesting uric acid-lowering drugs or HIF-1α inhibition as potential treatments for associated aortic valve calcification.
Collapse
Affiliation(s)
- Jialiang Zhang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, P.R. China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, P.R. China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Wenhua Lei
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, P.R. China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, P.R. China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jing Zhou
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, P.R. China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, P.R. China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Yaoyu Zhang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, P.R. China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, P.R. China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Fangyang Huang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, P.R. China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, P.R. China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, P.R. China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, P.R. China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
4
|
Blaser MC, Bäck M, Lüscher TF, Aikawa E. Calcific aortic stenosis: omics-based target discovery and therapy development. Eur Heart J 2025; 46:620-634. [PMID: 39656785 PMCID: PMC11825147 DOI: 10.1093/eurheartj/ehae829] [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: 07/19/2024] [Revised: 10/01/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Calcific aortic valve disease (CAVD) resulting in aortic stenosis (AS) is the most common form of valvular heart disease, affecting 2% of those over age 65. Those who develop symptomatic severe AS have an average further lifespan of <2 years without valve replacement, and three-quarters of these patients will develop heart failure, undergo valve replacement, or die within 5 years. There are no approved pharmaceutical therapies for AS, due primarily to a limited understanding of the molecular mechanisms that direct CAVD progression in the complex haemodynamic environment. Here, advances in efforts to understand the pathogenesis of CAVD and to identify putative drug targets derived from recent multi-omics studies [including (epi)genomics, transcriptomics, proteomics, and metabolomics] of blood and valvular tissues are reviewed. The recent explosion of single-cell omics-based studies in CAVD and the pathobiological and potential drug discovery insights gained from the application of omics to this disease area are a primary focus. Lastly, the translation of knowledge gained in valvular pathobiology into clinical therapies is addressed, with a particular emphasis on treatment regimens that consider sex-specific, renal, and lipid-mediated contributors to CAVD, and ongoing Phase I/II/III trials aimed at the prevention/treatment of AS are described.
Collapse
Affiliation(s)
- Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
| | - Magnus Bäck
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Valvular and Coronary Disease, Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 741, Boston, MA 02115, USA
| |
Collapse
|
5
|
Sivan S, Vijayakumar G, Pillai IC. Non-coding RNAs mediating the regulation of genes and signaling pathways in aortic valve calcification. Gene 2025; 936:149117. [PMID: 39580125 DOI: 10.1016/j.gene.2024.149117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Affiliation(s)
- Silpa Sivan
- Stem Cells and Regenerative Biology Lab, Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO, Kollam 690 525, Kerala, India
| | - Gayathri Vijayakumar
- Stem Cells and Regenerative Biology Lab, Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO, Kollam 690 525, Kerala, India
| | - Indulekha Cl Pillai
- Stem Cells and Regenerative Biology Lab, Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana PO, Kollam 690 525, Kerala, India.
| |
Collapse
|
6
|
Bartoli-Leonard F, Pennel T, Caputo M. Immunotherapy in the Context of Aortic Valve Diseases. Cardiovasc Drugs Ther 2024; 38:1173-1185. [PMID: 39017904 PMCID: PMC11680629 DOI: 10.1007/s10557-024-07608-7] [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] [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
|
7
|
Lan Y, Peng Q, Shen J, Liu H. Elucidating common biomarkers and pathways of osteoporosis and aortic valve calcification: insights into new therapeutic targets. Sci Rep 2024; 14:27827. [PMID: 39537712 PMCID: PMC11560947 DOI: 10.1038/s41598-024-78707-6] [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: 06/30/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Osteoporosis and aortic valve calcification, prevalent in the elderly, have unclear common mechanisms. This study aims to uncover them through bioinformatics analysis. METHODS Microarray data from GEO was analyzed for osteoporosis and aortic valve calcification. Differential expression analysis identified co-expressed genes. SVM-RFE and random forest selected key genes. GO and KEGG enrichment analyses were performed. Immunoinfiltration and GSEA analyses were subsequently performed. NetworkAnalyst analyzed microRNAs/TFs. HERB predicted drugs, and molecular docking assessed targeting potential. RESULTS Thirteen genes linked to osteoporosis and aortic valve calcification were identified. TNFSF11, KYNU, and HLA-DMB emerged as key genes. miRNAs, TFs, and drug predictions offered therapeutic insights. Molecular docking suggested 17-beta-estradiol and vitamin D3 as potential treatments. CONCLUSION The study clarifies shared mechanisms of osteoporosis and aortic valve calcification, identifies biomarkers, and highlights TNFSF11, KYNU, and HLA-DMB. It also suggests 17-beta-estradiol and vitamin D3 as potential effective treatments.
Collapse
Affiliation(s)
- Yujian Lan
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qingping Peng
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jianlin Shen
- Department of Orthopaedics, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China.
- Central Laboratory, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China.
| | - Huan Liu
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
8
|
Wei K, Cao Y, Kong X, Liu C, Gu X. Exploration and Validation of Immune and Therapeutic-Related Hub Genes in Aortic Valve Calcification and Carotid Atherosclerosis. J Inflamm Res 2024; 17:6485-6500. [PMID: 39310903 PMCID: PMC11416122 DOI: 10.2147/jir.s462546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Background Cardiovascular diseases, such as aortic valve calcification (AVC) and carotid atherosclerosis (CAS), impose substantial health challenges on a global scale. Both disorders have overlapping risk factors, which might trigger similar immune-inflammatory reactions in both diseases. Methods Shared differentially expressed genes (DEGs) were identified in the AVC and CAS datasets from the Gene Expression Omnibus (GEO). Candidate hub genes associated with immunity were identified using LASSO and immune cell infiltration analysis, and single gene set enrichment analysis (GSEA) was performed on the datasets. Subsequently, the hub genes were confirmed by qRT‒PCR validation in tissue samples. Results A total of 140 upregulated and 65 downregulated common genes were screened. Enrichment analyses highlighted immune system processes, response to stress, and cytokine pathways among the identified CEGs. LASSO identified candidate hub genes, including ANGPTL1, CX3CR1, and CCL4. Immune cell infiltration analysis emphasized the participation of immune cells, including macrophages, γδ T cells, and resting NK cells. The three hub genes were validated by qRT‒PCR analysis. Conclusion Our study explored immunological processes, including immune-related genes and cells, involved in the development of AVC and CAS. In particular, the identified hub genes ANGPTL1, CX3CR1, and CCL4 play crucial roles in mediating immune-inflammatory responses, which are central to the pathogenesis of these cardiovascular diseases, and the involvement of these genes in key immune pathways suggests that they could serve as potential biomarkers for early diagnosis or as targets for therapeutic strategies.
Collapse
Affiliation(s)
- KaiMing Wei
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yuan Cao
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - XiangJin Kong
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - ChuanZhen Liu
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - XingHua Gu
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
9
|
Oni E, Boakye E, Pressman GS, Dardari Z, Jha K, Szklo M, Budoff M, Nasir K, Hughes TM, Blaha MJ. The Association of Mitral Annular Calcification With Cardiovascular and Noncardiovascular Outcomes: The Multi-Ethnic Study of Atherosclerosis. Am J Cardiol 2024; 225:75-83. [PMID: 38914415 DOI: 10.1016/j.amjcard.2024.06.017] [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: 08/22/2023] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 06/26/2024]
Abstract
Mitral annular calcification (MAC) may be a potential marker of biologic aging. However, the association of MAC with noncardiovascular measures, including bone mineral density (BMD), incident renal failure, dementia, and noncardiovascular mortality, is not well-studied in a multiracial cohort. We used data from 6,814 participants (mean age: 62.2 ± 10.2 years, 52.9% women) without cardiovascular disease at baseline in the Multi-Ethnic Study of Atherosclerosis. MAC was assessed with noncontrast cardiac computed tomography at study baseline. Using multivariable-adjusted linear and logistic regression, we assessed the cross-sectional association of MAC with BMD and walking pace. Furthermore, using Cox proportional hazards, we evaluated the association of MAC with incident renal failure, dementia, and all-cause mortality. In addition, we assessed the association of MAC with cardiovascular and noncardiovascular mortality using competing risks regression. The prevalence of MAC was 9.5% and was higher in women (10.7%) than in men (8.0%). MAC was associated with low BMD (coefficient -0.04, 95% confidence interval [CI] -0.06 to -0.02), with significant interaction by gender (p-interaction = 0.035). MAC was, however, not associated with impaired walking pace (odds ratio 1.09, 95% CI 0.89 to 1.33). Compared with participants without MAC, those with MAC had an increased risk of incident renal failure, albeit nonsignificant (hazard ratio [HR] 1.18, 95% CI 0.95 to 1.45), and a significantly higher hazards of dementia (HR 1.36, 95% CI 1.10 to 1.70). In addition, participants with MAC had a substantially higher risk of all-cause (HR 1.47, 95% CI 1.29 to 1.69), cardiovascular (subdistribution HR 1.39, 95% CI 1.04 to 1.87), and noncardiovascular mortality (subdistribution HR 1.35, 95% CI 1.14 to 1.60) than those without MAC. MAC ≥100 versus <100 was significantly associated with reduced BMD, incident renal failure, dementia, all-cause, cardiovascular, and noncardiovascular mortality. In conclusion, MAC was associated with reduced BMD and dementia and all-cause, cardiovascular, and noncardiovascular mortality in this multiracial cohort. Thus, MAC may be a marker not only for atherosclerotic burden but also for other metabolic and inflammatory factors that increase the risk of noncardiovascular outcomes and death from other causes.
Collapse
Affiliation(s)
- Ebenezer Oni
- Division of Cardiology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania.
| | - Ellen Boakye
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gregg S Pressman
- Division of Cardiology, Einstein Medical Center, Philadelphia, Pennsylvania
| | - Zeina Dardari
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kunal Jha
- Division of Cardiovascular Medicine, University of Louisville, Louisville, Kentucky
| | - Moyses Szklo
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Matthew Budoff
- Lundquist Institute, Harbor-UCLA Medical Center, Torrance, California
| | - Khurram Nasir
- Division of Cardiovascular Prevention and Wellness, Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas
| | - Timothy M Hughes
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Michael J Blaha
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
10
|
Crosswhite P, Sun Z. TNFα Induces DNA and Histone Hypomethylation and Pulmonary Artery Smooth Muscle Cell Proliferation Partly via Excessive Superoxide Formation. Antioxidants (Basel) 2024; 13:677. [PMID: 38929115 PMCID: PMC11200563 DOI: 10.3390/antiox13060677] [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: 04/12/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Objective: The level of tumor necrosis factor-α (TNF-α) is upregulated during the development of pulmonary vascular remodeling and pulmonary hypertension. A hallmark of pulmonary arterial (PA) remodeling is the excessive proliferation of PA smooth muscle cells (PASMCs). The purpose of this study is to investigate whether TNF-α induces PASMC proliferation and explore the potential mechanisms. Methods: PASMCs were isolated from 8-week-old male Sprague-Dawley rats and treated with 0, 20, or 200 ng/mL TNF-α for 24 or 48 h. After treatment, cell number, superoxide production, histone acetylation, DNA methylation, and histone methylation were assessed. Results: TNF-α treatment increased NADPH oxidase activity, superoxide production, and cell numbers compared to untreated controls. TNF-α-induced PASMC proliferation was rescued by a superoxide dismutase mimetic tempol. TNF-α treatment did not affect histone acetylation at either dose but did significantly decrease DNA methylation. DNA methyltransferase 1 activity was unchanged by TNF-α treatment. Further investigation using QRT-RT-PCR revealed that GADD45-α, a potential mediator of DNA demethylation, was increased after TNF-α treatment. RNAi inhibition of GADD45-α alone increased DNA methylation. TNF-α impaired the epigenetic mechanism leading to DNA hypomethylation, which can be abolished by a superoxide scavenger tempol. TNF-α treatment also decreased H3-K4 methylation. TNF-α-induced PASMC proliferation may involve the H3-K4 demethylase enzyme, lysine-specific demethylase 1 (LSD1). Conclusions: TNF-α-induced PASMC proliferation may be partly associated with excessive superoxide formation and histone and DNA methylation.
Collapse
Affiliation(s)
- Patrick Crosswhite
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Human Physiology, Gonzaga University, Spokane, WA 99205, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| |
Collapse
|
11
|
Zhu Z, Liu Z, Zhang D, Li L, Pei J, Cai L. Models for calcific aortic valve disease in vivo and in vitro. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:6. [PMID: 38424219 PMCID: PMC10904700 DOI: 10.1186/s13619-024-00189-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Calcific Aortic Valve Disease (CAVD) is prevalent among the elderly as the most common valvular heart disease. Currently, no pharmaceutical interventions can effectively reverse or prevent CAVD, making valve replacement the primary therapeutic recourse. Extensive research spanning decades has contributed to the establishment of animal and in vitro cell models, which facilitates a deeper understanding of the pathophysiological progression and underlying mechanisms of CAVD. In this review, we provide a comprehensive summary and analysis of the strengths and limitations associated with commonly employed models for the study of valve calcification. We specifically emphasize the advancements in three-dimensional culture technologies, which replicate the structural complexity of the valve. Furthermore, we delve into prospective recommendations for advancing in vivo and in vitro model studies of CAVD.
Collapse
Affiliation(s)
- Zijin Zhu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China
| | - Zhirong Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China
| | - Li Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China.
| | - Jianqiu Pei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, China.
| | - Lin Cai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China.
| |
Collapse
|
12
|
Saji M, Nanasato M, Higuchi R, Izumi Y, Takamisawa I, Iguchi N, Shimizu J, Shimokawa T, Takayama M, Ikeda T, Isobe M. Impact of osteoporotic risk in women undergoing transcatheter aortic valve replacement. Cardiovasc Interv Ther 2024; 39:57-64. [PMID: 37231235 DOI: 10.1007/s12928-023-00940-z] [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: 02/01/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
Low body weight and advanced age are reported to be among the best predictors of osteoporosis, and osteoporosis self-assessment tool (OST) values are calculated using a simple formula to identify postmenopausal women at increased risk of osteoporosis. In our recent study, we demonstrated an association between fractures and poor outcomes in postmenopausal women following transcatheter aortic valve replacement (TAVR). In this study, we aimed to investigate the osteoporotic risk in women with severe aortic stenosis and determined whether an OST could predict all-cause mortality following TAVR. The study population comprised 619 women who underwent TAVR. Compared to a quarter of patients with diagnosis of osteoporosis, 92.4% of participants were at high risk of osteoporosis based on OST criteria. When divided into tertiles based on OST values, patients in tertile 1 (lowest OST) displayed increased frailty, a higher incidence of multiple fractures, and greater Society of Thoracic Surgeons scores. Estimated all-cause mortality survival rates 3 years post-TAVR were 84.2 ± 3.0%, 89.5 ± 2.6%, and 96.9 ± 1.7% for OST tertiles 1, 2, and 3, respectively (p = 0.001). Multivariate analysis showed that the OST tertile 3 was associated with decreased risk of all-cause mortality compared with OST tertile 1 as the referent. Notably, a history of osteoporosis was not associated with all-cause mortality. Patients with high osteoporotic risk are highly prevalent among those with aortic stenosis according to the OST criteria. OST value is a useful marker for predicting all-cause mortality in patients undergoing TAVR.
Collapse
Affiliation(s)
- Mike Saji
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, 6-11-1 Omori-nishi, Otaku, Tokyo, 143-8541, Japan.
- Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan.
| | - Mamoru Nanasato
- Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Ryosuke Higuchi
- Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Yuki Izumi
- Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Itaru Takamisawa
- Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Nobuo Iguchi
- Department of Cardiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Jun Shimizu
- Department of Anesthesiology, Sakakibara Heart Institute, Tokyo, Japan
| | - Tomoki Shimokawa
- Department of Cardiovascular Surgery, Sakakibara Heart Institute, Tokyo, Japan
| | | | - Takanori Ikeda
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, 6-11-1 Omori-nishi, Otaku, Tokyo, 143-8541, Japan
| | | |
Collapse
|
13
|
Szulc P, Lewis JR, Chapurlat R. Accelerated Bone Loss in Older Men With Severe Abdominal Aortic Calcification-the Prospective MINOS Study. J Clin Endocrinol Metab 2023; 109:e32-e39. [PMID: 37610245 DOI: 10.1210/clinem/dgad459] [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: 06/05/2023] [Revised: 06/30/2023] [Accepted: 08/07/2023] [Indexed: 08/24/2023]
Abstract
CONTEXT Data on the association between the severity of abdominal aortic calcification (AAC) and bone loss are discordant. OBJECTIVE Our aim was to assess the association between baseline AAC and prospectively assessed bone loss in older men. METHODS This prospective cohort study started in 1995 (MINOS). Men aged 50 to 85 years (n = 778) had AAC assessed on the lateral radiograph of the spine using Kauppila's semiquantitative score and was followed prospectively for 7.5 years. Bone mineral density (BMD) and bone mineral content (BMC) were measured by dual-energy x-ray absorptiometry every 18 months. Statistical analysis was performed using linear mixed models. RESULTS In comparison to men without AAC (AAC = 0), severe AAC (>6) was associated with more rapid bone loss at the total hip (-0.62 ± 0.06 vs -0.32 ± 0.04%/year; P < .001), trochanter, and distal forearm (-0.72 ± 0.06 vs -0.45 ± 0.03%/year; P < .001). The highest decile (AAC >10) was associated with more rapid bone loss at the femoral neck, whole body, and ultradistal radius (-0.86 ± 0.12 vs -0.34 ± 0.05%/year; P < .001). The results were similar for BMD and for BMC. The patterns were similar in sensitivity analyses (eg, after excluding men with abdominal obesity, after excluding current smokers, after excluding men with ischemic heart disease or with diabetes mellitus, after excluding men with abnormal concentrations of lipids, bioavailable 17β-estradiol or 25-hydroxycholecalciferol, after excluding men with glomerular filtration rate <60 mL/min). CONCLUSION Severe AAC is associated with faster bone loss in older men and may contribute to the higher fracture risk observed in this population.
Collapse
Affiliation(s)
- Pawel Szulc
- INSERM UMR 1033, University of Lyon, Hospices Civils de Lyon, 69437, Lyon, France
| | - Joshua R Lewis
- Institute for Nutrition Research, Edith Cowan University, Joondalup, Perth, WA 6027, Australia
- Medical School, the University of Western Australia, Perth, WA 6009, Australia
| | - Roland Chapurlat
- INSERM UMR 1033, University of Lyon, Hospices Civils de Lyon, 69437, Lyon, France
| |
Collapse
|
14
|
Bakhshian Nik A, Kaiser K, Sun P, Khomtchouk BB, Hutcheson JD. Altered Caveolin-1 Dynamics Result in Divergent Mineralization Responses in Bone and Vascular Calcification. Cell Mol Bioeng 2023; 16:299-308. [PMID: 37811003 PMCID: PMC10550882 DOI: 10.1007/s12195-023-00779-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/08/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction Though vascular smooth muscle cells adopt an osteogenic phenotype during pathological vascular calcification, clinical studies note an inverse correlation between bone mineral density and arterial mineral-also known as the calcification paradox. Both processes are mediated by extracellular vesicles (EVs) that sequester calcium and phosphate. Calcifying EV formation in the vasculature requires caveolin-1 (CAV1), a membrane scaffolding protein that resides in membrane invaginations (caveolae). Of note, caveolin-1-deficient mice, however, have increased bone mineral density. We hypothesized that caveolin-1 may play divergent roles in calcifying EV formation from vascular smooth muscle cells (VSMCs) and osteoblasts (HOBs). Methods Primary human coronary artery VSMCs and osteoblasts were cultured for up to 28 days in an osteogenic media. CAV1 expression was knocked down using siRNA. Methyl β-cyclodextrin (MβCD) and a calpain inhibitor were used, respectively, to disrupt and stabilize the caveolar domains in VSMCs and HOBs. Results CAV1 genetic variation demonstrates significant inverse relationships between bone-mineral density (BMD) and coronary artery calcification (CAC) across two independent epidemiological cohorts. Culture in osteogenic (OS) media increased calcification in HOBs and VSMCs. siRNA knockdown of CAV1 abrogated VSMC calcification with no effect on osteoblast mineralization. MβCD-mediated caveolae disruption led to a 3-fold increase of calcification in VSMCs treated with osteogenic media (p < 0.05) but hindered osteoblast mineralization (p < 0.01). Conversely, stabilizing caveolae by calpain inhibition prevented VSMC calcification (p < 0.05) without affecting osteoblast mineralization. There was no significant difference in CAV1 content between lipid domains from HOBs cultured in OS and control media. Conclusion Our data indicate fundamental cellular-level differences in physiological and pathophysiological mineralization mediated by CAV1 dynamics. This is the first study to suggest that divergent mechanisms in calcifying EV formation may play a role in the calcification paradox. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00779-7.
Collapse
Affiliation(s)
- Amirala Bakhshian Nik
- Department of Biomedical Engineering, Florida International University, 10555 W Flagler St, EC 2612, Miami, FL 33174 USA
| | - Katherine Kaiser
- Department of Biomedical Engineering, Florida International University, 10555 W Flagler St, EC 2612, Miami, FL 33174 USA
| | - Patrick Sun
- Department of BioHealth Informatics, Luddy School of Informatics, Computing, and Engineering, Indiana University, 535 W Michigan St, IT 477, Indianapolis, IN 46202 USA
| | - Bohdan B. Khomtchouk
- Department of BioHealth Informatics, Luddy School of Informatics, Computing, and Engineering, Indiana University, 535 W Michigan St, IT 477, Indianapolis, IN 46202 USA
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, IN USA
- Center for Computational Biology & Bioinformatics, Indiana University School of Medicine, Indianapolis, IN USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN USA
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, 10555 W Flagler St, EC 2612, Miami, FL 33174 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL USA
| |
Collapse
|
15
|
Chang GRL, Cheng WY, Fan HC, Chen HL, Lan YW, Chen MS, Yen CC, Chen CM. Kefir peptides attenuate atherosclerotic vascular calcification and osteoporosis in atherogenic diet-fed ApoE−/− knockout mice. Front Cell Dev Biol 2023; 11:1158812. [PMID: 37091976 PMCID: PMC10117689 DOI: 10.3389/fcell.2023.1158812] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/27/2023] [Indexed: 04/08/2023] Open
Abstract
Aims: Vascular calcification (VC) and osteoporosis were previously considered two distinct diseases. However, current understanding indicates that they share common pathogenetic mechanisms. The available medicines for treating VC and osteoporosis are limited. We previously demonstrated that kefir peptides (KPs) alleviated atherosclerosis in high-fat diet (HFD)-induced apolipoprotein E knockout (ApoE−/−) mice. The present study further addressed the preventive effects of KPs on VC and osteoporosis in ApoE−/− mice fed a high-cholesterol atherogenic diet (AD).Main methods: Seven-week-old ApoE−/− and wild-type C57BL/6 mice were randomly divided into five groups (n = 6). The development of VC and osteoporosis was evaluated after AD feeding for 13 weeks in KP-treated ApoE−/− mice and compared to C57BL/6 and ApoE−/− mice fed a standard chow diet (CD).Key findings: The results indicated that KP-treated ApoE−/− mice exhibited lower serum total cholesterol, oxidized low-density lipoprotein (ox-LDL), malondialdehyde (MDA) levels, and serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and creatine kinase (CK) activities, which suggested that KPs prevented hyperlipidemia and possible damages to the liver and muscle in ApoE−/− mice. KPs reduced serum tumor necrosis factor-α (TNF-α) and the local expression of TNF-α, IL-1β, and macrophage-specific CD68 markers in aortic tissues, which suggested that KPs inhibited inflammatory responses in AD-fed ApoE−/− mice. KPs reduced the deposition of lipid, collagen, and calcium minerals in the aortic roots of AD-fed ApoE−/− mice, which suggested that KPs inhibited the calcific progression of atherosclerotic plaques. KPs exerted osteoprotective effects in AD-fed ApoE−/− mice, which was evidenced by lower levels of the bone resorption marker CTX-1 and higher levels of the bone formation marker P1NP. KPs improved cortical bone mineral density and bone volume and reduced trabecular bone loss in femurs.Significance: The present data suggested that KPs attenuated VC and osteoporosis by reducing oxidative stress and inflammatory responses in AD-fed ApoE−/− mice. Our findings contribute to the application of KPs as preventive medicines for the treatment of hyperlipidemia-induced vascular and bone degeneration.
Collapse
Affiliation(s)
- Gary Ro-Lin Chang
- Department of Pediatrics, Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Taichung, Taiwan
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Yuan Cheng
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hueng-Chuen Fan
- Department of Pediatrics, Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Taichung, Taiwan
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Miaoli, Taiwan
| | - Hsiao-Ling Chen
- Department of Biomedical Sciences, and Department of Bioresources, Da-Yeh University, Changhwa, Taiwan
| | - Ying-Wei Lan
- Department of Pediatrics, Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Taichung, Taiwan
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Ming-Shan Chen
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Chih-Ching Yen
- Department of Internal Medicine, China Medical University Hospital, and College of Healthcare, China Medical University, Taichung, Taiwan
- *Correspondence: Chih-Ching Yen, ; Chuan-Mu Chen,
| | - Chuan-Mu Chen
- Department of Pediatrics, Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Taichung, Taiwan
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
- *Correspondence: Chih-Ching Yen, ; Chuan-Mu Chen,
| |
Collapse
|
16
|
Hadziselimovic E, Greve AM, Sajadieh A, Olsen MH, Kesäniemi YA, Nienaber CA, Ray SG, Rossebø AB, Wachtell K, Nielsen OW. Association of high-sensitivity troponin T with outcomes in asymptomatic non-severe aortic stenosis: a post-hoc substudy of the SEAS trial. EClinicalMedicine 2023; 58:101875. [PMID: 36915288 PMCID: PMC10006443 DOI: 10.1016/j.eclinm.2023.101875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND High-sensitivity Troponin T (hsTnT), a biomarker of cardiomyocyte overload and injury, relates to aortic valve replacement (AVR) and mortality in severe aortic stenosis (AS). However, its prognostic value remains unknown in asymptomatic patients with AS. We aimed to investigate if an hsTnT level >14 pg/mL (above upper limit of normal 99th percentile) is associated with echocardiographic AS-severity, subsequent AVR, ischaemic coronary events (ICE), and mortality in asymptomatic patients with non-severe AS. METHODS In this post-hoc sub-analysis of the multicentre, randomised, double-blind, placebo-controlled SEAS trial (ClinicalTrials.gov, NCT00092677), we included asymptomatic patients with mild to moderate-severe AS. We ascertained baseline and 1-year hsTnT concentrations and examined the association between baseline levels and the risk of the primary composite endpoint, defined as the first event of all-cause mortality, isolated AVR (without coronary artery bypass grafting (CABG)), or ICE. Multivariable regressions and competing risk analyses examined associations of hsTnT level >14 pg/mL with clinical correlates and 5-year risk of the primary endpoint. FINDINGS Between January 6, 2003, and March 4, 2004, a total of 1873 patients were enrolled in the SEAS trial, and 1739 patients were included in this post-hoc sub-analysis. Patients had a mean (SD) age of 67.5 (9.7) years, 61.0% (1061) were men, 17.4% (302) had moderate-severe AS, and 26.0% (453) had hsTnT level >14 pg/mL. The median hsTnT difference from baseline to 1-year was 0.8 pg/mL (IQR, -0.4 to 2.3). In adjusted linear regression, log(hsTnT) did not correlate with echocardiographic AS severity (p = 0.36). In multivariable Cox regression, a hsTnT level >14 pg/mL vs. hsTnT ≤14 pg/mL was associated with an increased risk of the primary composite endpoint (HR, 1.41; 95% CI, 1.18-1.70; p = 0.0002). In a competing risk model of first of the individual components of the primary endpoint, a hsTnT level >14 pg/mL was associated with ICE risk (HR 1.71; 95% CI, 1.23-2.38; p = 0.0013), but not with isolated AVR (p = 0.064) or all-cause mortality (p = 0.49) as the first event. INTERPRETATION hsTnT level is within the reference range (≤14 pg/mL) in 3 out of 4 non-ischaemic patients with asymptomatic mild-to-moderate AS and remains stable during a 1-year follow-up regardless of AS-severity. An hsTnT level >14 pg/mL was mainly associated with subsequent ICE, which suggest that hsTnT concentration is primarily a risk marker of subclinical coronary atherosclerotic disease. FUNDING Merck & Co., Inc., the Schering-Plough Corporation, the Interreg IVA program, Roche Diagnostics Ltd., and Gangstedfonden. Open access publication fee funding provided by prof. Olav W. Nielsen and Department of Cardiology, Bispebjerg University Hospital, Denmark.
Collapse
Affiliation(s)
- Edina Hadziselimovic
- Department of Cardiology, Bispebjerg University Hospital, Copenhagen, Denmark
- Corresponding author. Department of Cardiology, Bispebjerg University Hospital, Bispebjerg Bakke 23, 2400, Copenhagen, Denmark.
| | - Anders M. Greve
- Department of Clinical Biochemistry, 3011, Rigshospitalet, Copenhagen, Denmark
| | - Ahmad Sajadieh
- Department of Cardiology, Bispebjerg University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Michael H. Olsen
- Department of Internal Medicine 1, Holbæk Hospital, Denmark
- Department of Regional Health Research, University of Southern Denmark, Denmark
| | - Y. Antero Kesäniemi
- Research Unit of Internal Medicine, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | | | | | - Anne B. Rossebø
- Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway
| | | | - Olav W. Nielsen
- Department of Cardiology, Bispebjerg University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
17
|
Wen D, Hu L, Shan J, Zhang H, Hu L, Yuan A, Pu J, Xue S. Mechanical injury accentuates lipid deposition in ApoE -/- mice and advance aortic valve stenosis: A novel modified aortic valve stenosis model. Front Cardiovasc Med 2023; 10:1119746. [PMID: 36818346 PMCID: PMC9932047 DOI: 10.3389/fcvm.2023.1119746] [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: 12/09/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Background Current mouse models still have limitations in studying aortic valve stenosis (AVS). A suitable animal model bearing a close resemblance to the pathophysiological processes of humans needs to be developed. Here, we combined two risk factors to create a mouse model that mimics the pathological features of human AVS. Methods and results We combined WI and hyperlipidemia in ApoE-/- mice to explore the synergistic effect on the stenosis of the aortic valve. Transthoracic echocardiography revealed progressively increased peak velocity with age in ApoE-/- mice to velocities above C57 mice when fed a high-fat diet after wire injury. Moreover, ApoE-/- mice demonstrated lower cusp separation and lower aortic valve area after 8 weeks vs. C57 mice. Gross morphology and MRI showed advanced thickening, sclerosis aortic valve, narrowing of the orifice area, and micro-CT showed obvious calcification in the aortic valves in the hyperlipidemia group after wire injury. Histopathology studies showed thickening and fibrosis of aortic valve leaflets in the hyperlipidemia group after wire injury. Notably, lipid deposition was observed in ApoE-/- mice 8 weeks after wire injury, accompanied by overexpressed apoB and apoA proteins. After wire injury, the hyperlipidemia group exhibited augmented inflammation, ROS production, and apoptosis in the leaflets. Moreover, the combination group exhibited advanced fibro-calcific aortic valves after wire injury. Conclusion Overall, we present the synergistic effect of wire injury and hyperlipidemia on lipoproteins deposition in the development of AVS in ApoE-/- mice, this model bear close resemblance to human AVS pathology.
Collapse
Affiliation(s)
- Dezhong Wen
- Department of Cardiovascular Surgery, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Hu
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianggui Shan
- Department of Cardiovascular Surgery, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengyuan Zhang
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liuhua Hu
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ancai Yuan
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Key Laboratory of Coronary Heart Disease, Shanghai Municipal Education Commission, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Jun Pu,
| | - Song Xue
- Department of Cardiovascular Surgery, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Song Xue,
| |
Collapse
|
18
|
Turner ME, Bartoli‐Leonard F, Aikawa E. Small particles with large impact: Insights into the unresolved roles of innate immunity in extracellular vesicle‐mediated cardiovascular calcification. Immunol Rev 2022; 312:20-37. [DOI: 10.1111/imr.13134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mandy E Turner
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Francesca Bartoli‐Leonard
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
- Division of Cardiovascular Medicine Department of Medicine Center for Excellence in Vascular Biology Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| |
Collapse
|
19
|
Maternal high-fat diet promotes calcified atherosclerotic plaque formation in adult offspring by enhancing transformation of VSMCs to osteochondrocytic-like phenotype. Heliyon 2022; 8:e10644. [PMID: 36158105 PMCID: PMC9489965 DOI: 10.1016/j.heliyon.2022.e10644] [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: 11/29/2021] [Revised: 03/17/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
Aim Maternal high-fat diet (HFD) is associated with the development of cardiovascular disease (CVD) in adult offspring. Atherosclerotic vascular calcification is well documented in patients with CVD. We examined the effect of maternal HFD on calcified plaque formation. Methods and results Seven-week-old female apo-E−/− mice (C57BL6/J) were nourished either an HFD or a normal diet (ND) a week before mating, and during gestation and lactation. Offspring of both the groups were fed a high-cholesterol diet (HCD) from 8 weeks of age. Osteogenic activity of the thoracic aorta, assessed using an ex vivo imaging system, was significantly increased after 3 months of HCD in male offspring of HFD-fed dams (O-HFD) as compared with those of ND-fed dams (O-ND). Alizarin-red-positive area in the aortic root was significantly increased after 6 months of HCD in male O-HFD as compared to that of O-ND. Plaque size and Oil Red O-positive staining were comparable between the two groups. Primary cultured vascular smooth muscle cells (VSMCs) of the thoracic aorta were treated with phosphate and interleukinL-1β (IL-1β) to transform them into an osteochondrocytic-like phenotype. Intracellular calcium content and alkaline phosphatase activity were markedly higher in the VSMCs of O-HFD than in O-ND. IL-1β concentration in the supernatant of bone marrow-derived macrophages was markedly higher in O-HFD than in O-ND. Conclusion Our findings indicate that maternal HFD accelerates the expansion of atherogenic calcification independent of plaque progression. In vitro phosphate- and IL-1β-induced osteochondrocytic transformation of VSMCs was augmented in O-HFD. Inhibition of VSMCs, skewing toward osteochondrocytic-like cells, might be a potential therapeutic strategy for preventing maternal HFD-associated CVD development.
Collapse
|
20
|
Extracellular Vesicles, Inflammation, and Cardiovascular Disease. Cells 2022; 11:cells11142229. [PMID: 35883672 PMCID: PMC9320258 DOI: 10.3390/cells11142229] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. The underlying mechanisms of most cardiovascular disorders involve innate and adaptive immune responses, and extracellular vesicles are implicated in both. In this review, we describe the mechanistic role of extracellular vesicles at the intersection of inflammatory processes and cardiovascular disease. Our discussion focuses on atherosclerosis, myocardial ischemia and ischemic heart disease, heart failure, aortic aneurysms, and valvular pathology.
Collapse
|
21
|
Lu J, Xie S, Deng Y, Xie X, Liu Y. Blocking the NLRP3 inflammasome reduces osteogenic calcification and M1 macrophage polarization in a mouse model of calcified aortic valve stenosis. Atherosclerosis 2022; 347:28-38. [DOI: 10.1016/j.atherosclerosis.2022.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/02/2022] [Accepted: 03/02/2022] [Indexed: 02/06/2023]
|
22
|
Matsui M, Bouchareb R, Storto M, Hussain Y, Gregg A, Marx SO, Pitt GS. Increased Ca2+ influx through CaV1.2 drives aortic valve calcification. JCI Insight 2022; 7:155569. [PMID: 35104251 PMCID: PMC8983132 DOI: 10.1172/jci.insight.155569] [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: 10/05/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is heritable as revealed by recent genome wide association studies. While polymorphisms linked to increased expression of CACNA1C, encoding the CaV1.2 L-type voltage-gated Ca2+ channel, and increased Ca2+ signaling are associated with CAVD, whether increased Ca2+ influx through the druggable CaV1.2 is causal for calcific aortic valve disease is unknown. With surgically removed aortic valves from patients, we confirmed the association between increased CaV1.2 expression and CAVD. We extended our studies with a transgenic mouse model that mimics increased CaV1.2 expression in within aortic valve interstitial cells (VICs). In young mice maintained on normal chow, we observed dystrophic valve lesions that mimic changes found in pre-symptomatic CAVD, and showed activation of chondrogenic and osteogenic transcriptional regulators within these valve lesions. Chronic administration of verapamil, a clinically used CaV1.2 antagonist, slowed the progression of lesion development in vivo. Exploiting VIC cultures we demonstrated that increased Ca2+ influx through CaV1.2 drives signaling programs that lead to myofibroblast activation of VICs and upregulation of genes associated with aortic valve calcification. Our data support a causal role for Ca2+ influx through CaV1.2 in CAVD and suggest that early treatment with Ca2+ channel blockers is an effective therapeutic strategy.
Collapse
Affiliation(s)
- Maiko Matsui
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| | - Rihab Bouchareb
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Mara Storto
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| | - Yasin Hussain
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| | - Andrew Gregg
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, United States of America
| | - Geoffrey S Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, United States of America
| |
Collapse
|
23
|
Zhou Q, Cao H, Hang X, Liang H, Zhu M, Fan Y, Shi J, Dong N, He X. Midkine Prevents Calcification of Aortic Valve Interstitial Cells via Intercellular Crosstalk. Front Cell Dev Biol 2022; 9:794058. [PMID: 34977035 PMCID: PMC8714929 DOI: 10.3389/fcell.2021.794058] [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: 10/13/2021] [Accepted: 11/24/2021] [Indexed: 12/17/2022] Open
Abstract
Calcified aortic valve disease (CAVD), the most common valvular heart disease, lacks pharmaceutical treatment options because its pathogenesis remains unclear. This disease with a complex macroenvironment characterizes notable cellular heterogeneity. Therefore, a comprehensive understanding of cellular diversity and cell-to-cell communication are essential for elucidating the mechanisms driving CAVD progression and developing therapeutic targets. In this study, we used single-cell RNA sequencing (scRNA-seq) analysis to describe the comprehensive transcriptomic landscape and cell-to-cell interactions. The transitional valvular endothelial cells (tVECs), an intermediate state during the endothelial-to-mesenchymal transition (EndMT), could be a target to interfere with EndMT progression. Moreover, matrix valvular interstitial cells (mVICs) with high expression of midkine (MDK) interact with activated valvular interstitial cells (aVICs) and compliment-activated valvular interstitial cells (cVICs) through the MK pathway. Then, MDK inhibited calcification of VICs that calcification was validated by Alizarin Red S staining, real-time quantitative polymerase chain reaction (RT-qPCR), and Western blotting assays in vitro. Therefore, we speculated that mVICs secreted MDK to prevent VICs’ calcification. Together, these findings delineate the aortic valve cells’ heterogeneity, underlining the importance of intercellular cross talk and MDK, which may offer a potential therapeutic strategy as a novel inhibitor of CAVD.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyi Hang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Huamin Liang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Miaomiao Zhu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Yixian Fan
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ximiao He
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
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: 37] [Impact Index Per Article: 9.3] [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
|
25
|
Abstract
PURPOSE OF REVIEW Calcific aortic stenosis (CAVS) is the most common form of valvular heart disease in developed countries, increasing in prevalence with the aging population. Surgical or transcatheter aortic valve replacement is the only treatment available for CAVS. However, these interventions are typically reserved for severe symptomatic aortic stenosis (AS). The purpose of this review is to summarize the recent literature in uncovering the underlying pathophysiology of CAVS in the setting of lipoprotein (a) [Lp(a)] and emerging therapies targeting Lp(a) which may help halt disease progression in CAVS. RECENT FINDINGS Pathophysiologic, epidemiological, and genetic studies over the past two decades have provided strong evidence that Lp(a) is an important mediator of calcific aortic valvular disease (CAVD). Studies suggest that Lp(a) is a key carrier of pro-calcifying oxidized phospholipids (OxPL). The metabolism of OxPL results in a pro-inflammatory state and subsequent valvular thickening and mineralization through pro-osteogenic signaling. The identification of Lp(a) as a causal mediator of CAVD has allowed for opportunities for emerging therapeutic agents which may slow the progression of CAVD (Fig. 1JOURNAL/cocar/04.03/00001573-202109000-00007/figure1/v/2021-08-04T080204Z/r/image-jpeg). SUMMARY This review summarizes the current knowledge on the association of Lp(a) with CAVD and ongoing studies of potential Lp(a)-lowering therapies. Based on the rate-limiting and causal role of Lp(a) in progression of CAVS, these therapies may represent novel pharmacotherapies in AS and inform the developing role of Lp(a) in the clinical management of CAVD.
Collapse
|
26
|
Vinchi F. Non-Transferrin-Bound Iron in the Spotlight: Novel Mechanistic Insights into the Vasculotoxic and Atherosclerotic Effect of Iron. Antioxid Redox Signal 2021; 35:387-414. [PMID: 33554718 PMCID: PMC8328045 DOI: 10.1089/ars.2020.8167] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/06/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
Significance: While atherosclerosis is an almost inevitable consequence of aging, food preferences, lack of exercise, and other aspects of the lifestyle in many countries, the identification of new risk factors is of increasing importance to tackle a disease, which has become a major health burden for billions of people. Iron has long been suspected to promote the development of atherosclerosis, but data have been conflicting, and the contribution of iron is still debated controversially. Recent Advances: Several experimental and clinical studies have been recently published about this longstanding controversial problem, highlighting the critical need to unravel the complexity behind this topic. Critical Issues: The aim of the current review is to provide an overview of the current knowledge about the proatherosclerotic impact of iron, and discuss the emerging role of non-transferrin-bound iron (NTBI) as driver of vasculotoxicity and atherosclerosis. Finally, I will provide detailed mechanistic insights on the cellular processes and molecular pathways underlying iron-exacerbated atherosclerosis. Overall, this review highlights a complex framework where NTBI acts at multiple levels in atherosclerosis by altering the serum and vascular microenvironment in a proatherogenic and proinflammatory manner, affecting the functionality and survival of vascular cells, promoting foam cell formation and inducing angiogenesis, calcification, and plaque destabilization. Future Directions: The use of additional iron markers (e.g., NTBI) may help adequately predict predisposition to cardiovascular disease. Clinical studies are needed in the aging population to address the atherogenic role of iron fluctuations within physiological limits and the therapeutic value of iron restriction approaches. Antioxid. Redox Signal. 35, 387-414.
Collapse
Affiliation(s)
- Francesca Vinchi
- Iron Research Program, Lindsley F. Kimball Research Institute (LFKRI), New York Blood Center (NYBC), New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| |
Collapse
|
27
|
Fan Y, Shao J, Wei S, Song C, Li Y, Jiang S. Self-eating and Heart: The Emerging Roles of Autophagy in Calcific Aortic Valve Disease. Aging Dis 2021; 12:1287-1303. [PMID: 34341709 PMCID: PMC8279526 DOI: 10.14336/ad.2021.0101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/01/2021] [Indexed: 12/16/2022] Open
Abstract
Autophagy is a self-degradative pathway by which subcellular elements are broken down intracellularly to maintain cellular homeostasis. Cardiac autophagy commonly decreases with aging and is accompanied by the accumulation of misfolded proteins and dysfunctional organelles, which are undesirable to the cell. Reduction of autophagy over time leads to aging-related cardiac dysfunction and is inversely related to longevity. However, despite the increasing interest in autophagy in cardiac diseases and aging, the process remains an undervalued and disregarded object in calcific valvular disease. Neither the nature through which autophagy is triggered nor the interplay between autophagic machinery and targeted molecules during aortic valve calcification are fully understood. Recently, the upregulation of autophagy has been shown to result in cardioprotective effects against cell death as well as its origin. Here, we review the evidence that shows how autophagy can be both beneficial and detrimental as it pertains to aortic valve calcification in the heart.
Collapse
Affiliation(s)
- Yunlong Fan
- Medical School of Chinese PLA, Beijing 100853, China.
- Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Jiakang Shao
- Medical School of Chinese PLA, Beijing 100853, China.
| | - Shixiong Wei
- Medical School of Chinese PLA, Beijing 100853, China.
- Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Song
- Medical School of Chinese PLA, Beijing 100853, China.
- Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Yanan Li
- Medical School of Chinese PLA, Beijing 100853, China.
| | - Shengli Jiang
- Medical School of Chinese PLA, Beijing 100853, China.
- Department of Cardiovascular Surgery, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
28
|
Xie B, Chen S, Xu Y, Han W, Hu R, Chen M, Zhang Y, Ding S. The Impact of Glucagon-Like Peptide 1 Receptor Agonists on Bone Metabolism and Its Possible Mechanisms in Osteoporosis Treatment. Front Pharmacol 2021; 12:697442. [PMID: 34220521 PMCID: PMC8243369 DOI: 10.3389/fphar.2021.697442] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus and osteoporosis are closely related and have complex influencing factors. The impact of anti-diabetic drugs on bone metabolism has received more and more attention. Type 2 diabetes mellitus (T2DM) would lead to bone fragility, high risk of fracture, poor bone repair and other bone-related diseases. Furthermore, hypoglycemic drugs used to treat T2DM may have notable detrimental effects on bones. Thus, the clinically therapeutic strategy for T2DM should not only effectively control the patient's glucose levels, but also minimize the complications of bone metabolism diseases. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are novel and promising drug for the treatment of T2DM. Some studies have found that GLP-1RAs may play an anti-osteoporotic effect by controlling blood sugar levels, promoting bone formation and inhibiting bone resorption. However, in clinical practice, the specific effects of GLP-1RA on fracture risk and osteoporosis have not been clearly defined and evidenced. This review summarizes the current research findings by which GLP-1RAs treatment of diabetic osteoporosis, postmenopausal osteoporosis and glucocorticoid-induced osteoporosis and describes possible mechanisms, such as GLP-1R/MAPK signaling pathway, GLP-1R/PI3K/AKT signaling pathway and Wnt/β-catenin pathway, that are associated with GLP-1RAs and osteoporosis. The specific role and related mechanisms of GLP-1RAs in the bone metabolism of patients with different types of osteoporosis need to be further explored and clarified.
Collapse
Affiliation(s)
- Baocheng Xie
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Shichun Chen
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yongxiang Xu
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Weichao Han
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Runkai Hu
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Minyi Chen
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Yusheng Zhang
- Department of Pharmacy, The First People's Hospital of Foshan (The Affiliated Foshan Hospital of Sun Yat-Sen University), Foshan, China
| | - Shaobo Ding
- Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| |
Collapse
|
29
|
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: 0.5] [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
|
30
|
Sia SK, Jan MS, Wang YH, Huang YF, Wei JCC. Periodontitis is associated with incidental valvular heart disease: A nationwide population-based cohort study. J Clin Periodontol 2021; 48:1085-1092. [PMID: 34060116 DOI: 10.1111/jcpe.13478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/18/2021] [Accepted: 04/10/2021] [Indexed: 12/15/2022]
Abstract
AIM Periodontitis and valvular heart disease (VHD) are common diseases. Both diseases are related to chronic inflammation and share many common risk factors. Previous periodontal studies had focused mainly on atherosclerotic cardiovascular disease. This study aimed to determine whether periodontitis is associated with the development of VHD. MATERIALS AND METHODS This was a retrospective nationwide cohort study using Taiwan's Longitudinal Health Insurance Database. Using ICD-9-CM coding, both the periodontitis and non-periodontitis groups were matched. RESULTS There were 8483 cases and 4919 cases of VHD diagnosed in the periodontitis group and non-periodontitis group, respectively. The cumulative incidence of VHD was significantly higher in the periodontitis group (log-rank test, p < .001), with the incidence density of 6.44 (95% CI, 6.31-6.58) per 1000 person-years in the periodontitis group compared to 4.65 (95% CI, 4.52-4.78) in the non-periodontitis group. The relative risk for VHD was 1.39 (95% CI, 1.34-1.44). After multivariate analysis, periodontitis was independently associated with a risk for VHD (HR, 1.38; 95% CI, 1.33-1.42, p < .001). Intensive treatment of periodontitis significantly lowered the risk for VHD (HR, 0.68; 95% CI, 0.60-0.77, p < .001). CONCLUSIONS Periodontitis was significantly associated with the development of VHD. Treatment of periodontitis reduced the risk for VHD.
Collapse
Affiliation(s)
- Sung-Kien Sia
- Division of cardiology, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Shiou Jan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Feng Huang
- Department of stomatology, Chung Shan Medical University Hospital, Taichung, Taiwan.,College of oral medicine, Chung Shan Medical University, Taichung, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan.,Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
31
|
Wang Y, Weng Y, Li X, Huang Q, Xiang Y, Li X, Shi Q. Dihydrotanshinone I inhibits aortic valve interstitial cell calcification via the SMAD1/5/8/NF-κB/ERK pathway. Biomed Pharmacother 2021; 139:111674. [PMID: 34243610 DOI: 10.1016/j.biopha.2021.111674] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVES In calcific aortic valve disease (CAVD), the valve interstitial cells (VIC) osteogenic phenotype changes can lead to thickening and calcification of the valve leaflets,eventually lead to restricted valve movement and life-threatening. This study aims to investigate the effect and mechanism of dihydrotanshinone I (DHI) on osteogenic medium (OM) induced osteogenic phenotypic transition of porcine valve interstitial cells (PVICs), which can provide theoretical and scientific basis for clinical intervention in CAVD. METHODS AND RESULTS Immunohistochemical methods were used to detect the expression of osteogenic indicators Runx2, OPN and inflammation indicators IL-1β and p-NF-κB in valve specimens of CAVD patients(N = 3) and normal controls(N = 1). PVICs stimulated by osteoblastic medium (OM) were treated with or without DHI. CCK8, ALP and Alizarin Red S staining were used to detect cell growth and calcification, respectively. The results showed that under the treated with DHI, compared with OM, the formation of calcium nodules was reduced, and the expression of calcification-related markers Runx2 and OPN were down-regulated, which quantified by qRT-PCR and western blot. In addition, on the basis of OM induction, DHI also inhibited the phosphorylation of the NF-κB/ERK1/2 and SMAD1/5/8 signaling pathway. CONCLUSION DHI (10 μM) treatment can reverse the osteogenic phenotypic transition of PVICs induced by osteogenic medium, and the mechanism may be related to NF-κB、ERK 1/2 and Smad1/5/8 pathways.
Collapse
Affiliation(s)
- Yue Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yaguang Weng
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xian Li
- Department of Pathology, Chongqing Medical University, China
| | - Qin Huang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yi Xiang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaorong Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Qiong Shi
- Ministry of Education Key Laboratory of Diagnostic Medicine, and School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
32
|
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: 47] [Impact Index Per Article: 11.8] [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
|
33
|
Nakamizo T, Cologne J, Cordova K, Yamada M, Takahashi T, Misumi M, Fujiwara S, Matsumoto M, Kihara Y, Hida A, Ohishi W. Radiation effects on atherosclerosis in atomic bomb survivors: a cross-sectional study using structural equation modeling. Eur J Epidemiol 2021; 36:401-414. [PMID: 33742296 PMCID: PMC8076141 DOI: 10.1007/s10654-021-00731-x] [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: 09/04/2020] [Accepted: 02/10/2021] [Indexed: 12/24/2022]
Abstract
Past reports indicated that total-body irradiation at low to moderate doses could be responsible for cardiovascular disease risks, but the mechanism remains unclear. The purpose of this study was to investigate the association between radiation exposure and atherosclerosis, an underlying pathology of cardiovascular diseases, in the Japanese atomic bomb survivors. We performed a cross-sectional study measuring 14 clinical-physiological atherosclerosis indicators during clinical exams from 2010 to 2014 in 3274 participants of the Adult Health Study cohort. Multivariable analyses were performed by using a structural equation model with latent factors representing underlying atherosclerotic pathologies: (1) arterial stiffness, (2) calcification, and (3) plaque as measured with indicators chosen a priori on the basis of clinical-physiological knowledge. Radiation was linearly associated with calcification (standardized coefficient per Gy 0.15, 95 % confidence interval: CI [0.070, 0.23]) and plaque (0.11, 95 % CI [0.029, 0.20]), small associations that were comparable to about 2 years of aging per Gy of radiation exposure, but not with arterial stiffness (0.036, 95 % CI [− 0.025, 0.095]). The model fitted better and had narrower confidence intervals than separate ordinary regression models explaining individual indicators independently. The associations were less evident when the dose range was restricted to a maximum of 2 or 1 Gy. By combining individual clinical-physiological indicators that are correlated because of common, underlying atherosclerotic pathologies, we found a small, but significant association of radiation with atherosclerosis.
Collapse
Affiliation(s)
- Tomoki Nakamizo
- Department of Clinical Studies, Radiation Effects Research Foundation (RERF), Nagasaki, Japan.
| | - John Cologne
- Department of Statistics, RERF, Hiroshima, Japan
| | | | | | - Tetsuya Takahashi
- Faculty of Rehabilitation, Hiroshima International University, Hiroshima, Japan
| | | | - Saeko Fujiwara
- Department of Clinical Studies, RERF, Hiroshima, Japan.,Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan
| | - Masayasu Matsumoto
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University, Hiroshima, Japan.,Department of Neurology, Sakai City Medical Center, Osaka, Japan
| | - Yasuki Kihara
- Department of Clinical Studies, RERF, Hiroshima, Japan.,Department of Cardiovascular Medicine, Hiroshima University, Hiroshima, Japan.,Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Ayumi Hida
- Department of Clinical Studies, Radiation Effects Research Foundation (RERF), Nagasaki, Japan
| | - Waka Ohishi
- Department of Clinical Studies, RERF, Hiroshima, Japan
| |
Collapse
|
34
|
Kim JM, Park KY, Kim HR, Ahn HY, Pantoni L, Park MS, Han SH, Jung HB, Bae J. Association of Bone Mineral Density to Cerebral Small Vessel Disease Burden. Neurology 2021; 96:e1290-e1300. [PMID: 33431517 DOI: 10.1212/wnl.0000000000011526] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/10/2020] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To test the hypothesis that bone mineral loss is mechanistically related to cerebral small vessel disease (SVD), we investigated the relationship between bone mineral density and the prevalence and intensity of SVD among patients with stroke. METHODS We analyzed data of 1,190 consecutive patients with stroke who were >50 years of age and underwent both brain MRI and dual-energy x-ray absorptiometry from the stroke registry of Chung-Ang University Hospital in Seoul, Korea. The patients were categorized into 3 groups according to their bone mineral density (normal, osteopenia, and osteoporosis). White matter hyperintensities, silent lacunes, cerebral microbleeds, and extensive perivascular space were assessed from brain MRI. Multinomial logistic regression model was used to examine the association between osteoporosis and total SVD score. We also recruited 70 patients with stroke to study serum bone turnover markers and microRNAs related to both cerebral atherosclerosis and bone metabolism to understand bone and brain interaction. RESULTS Osteoporosis was determined among 284 patients (23.9%), and 450 patients (37.8%) had osteopenia. As bone mineral density decreased, total SVD score and the incidence of every SVD phenotype increased except strictly lobar cerebral microbleeds. Multinomial logistic regression analysis showed that osteoporosis was independently associated with severe SVD burden. The levels of microRNA-378f were significantly increased among the patients with osteoporosis and maximal total SVD score and positively correlated with parathyroid hormone and osteocalcin. CONCLUSIONS These findings suggest a pathophysiologic link between bone mineral loss and hypertensive cerebral arteriolar degeneration, possibly mediated by circulating microRNA.
Collapse
Affiliation(s)
- Jeong-Min Kim
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea
| | - Kwang-Yeol Park
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea.
| | - Hye Ryoun Kim
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea
| | - Hwa Young Ahn
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea
| | - Leonardo Pantoni
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea
| | - Moo-Seok Park
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea.
| | - Su-Hyun Han
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea
| | - Hae-Bong Jung
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea
| | - Jaehan Bae
- From the Department of Neurology (J.-M.K.), Seoul National University Hospital; Departments of Neurology (K.-Y.P., S.-H.H., H.-B.J., J.B.), Laboratory Medicine (H.R.K.), and Internal Medicine (H.Y.A.), Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea; Luigi Sacco Stroke and Dementia Lab (L.P.), Department of Biomedical and Clinical Sciences, University of Milan, Italy; and Department of Neurology (M.-S.P.), Seoul Hospital Ewha Womans University College of Medicine, South Korea
| |
Collapse
|
35
|
Grim JC, Aguado BA, Vogt BJ, Batan D, Andrichik CL, Schroeder ME, Gonzalez-Rodriguez A, Yavitt FM, Weiss RM, Anseth KS. Secreted Factors From Proinflammatory Macrophages Promote an Osteoblast-Like Phenotype in Valvular Interstitial Cells. Arterioscler Thromb Vasc Biol 2020; 40:e296-e308. [PMID: 32938214 DOI: 10.1161/atvbaha.120.315261] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Resident valvular interstitial cells (VICs) activate to myofibroblasts during aortic valve stenosis progression, which further promotes fibrosis or even differentiate into osteoblast-like cells that can lead to calcification of valve tissue. Inflammation is a hallmark of aortic valve stenosis, so we aimed to determine proinflammatory cytokines secreted from M1 macrophages that give rise to a transient VIC phenotype that leads to calcification of valve tissue. Approach and Results: We designed hydrogel biomaterials as valve extracellular matrix mimics enabling the culture of VICs in either their quiescent fibroblast or activated myofibroblast phenotype in response to the local matrix stiffness. When VIC fibroblasts and myofibroblasts were treated with conditioned media from THP-1-derived M1 macrophages, we observed robust reduction of αSMA (alpha smooth muscle actin) expression, reduced stress fiber formation, and increased proliferation, suggesting a potent antifibrotic effect. We further identified TNF (tumor necrosis factor)-α and IL (interleukin)-1β as 2 cytokines in M1 media that cause the observed antifibrotic effect. After 7 days of culture in M1 conditioned media, VICs began differentiating into osteoblast-like cells, as measured by increased expression of RUNX2 (runt-related transcription factor 2) and osteopontin. We also identified and validated IL-6 as a critical mediator of the observed pro-osteogenic effect. CONCLUSIONS Proinflammatory cytokines in M1 conditioned media inhibit myofibroblast activation in VICs (eg, TNF-α and IL-1β) and promote their osteogenic differentiation (eg, IL-6). Together, our work suggests inflammatory M1 macrophages may drive a myofibroblast-to-osteogenic intermediate VIC phenotype, which may mediate the switch from fibrosis to calcification during aortic valve stenosis progression.
Collapse
Affiliation(s)
- Joseph C Grim
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Brian A Aguado
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Brandon J Vogt
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Dilara Batan
- BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,Division of Biochemistry (D.B.), University of Colorado Boulder, Boulder
| | - Cassidy L Andrichik
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Megan E Schroeder
- BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,Materials Science and Engineering Program (M.E.S., K.S.A.), University of Colorado Boulder, Boulder
| | - Andrea Gonzalez-Rodriguez
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - F Max Yavitt
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Robert M Weiss
- Department of Internal Medicine, University of Iowa, Iowa City (R.M.W.)
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,Materials Science and Engineering Program (M.E.S., K.S.A.), University of Colorado Boulder, Boulder
| |
Collapse
|
36
|
Tap L, Kirkham FA, Mattace-Raso F, Joly L, Rajkumar C, Benetos A. Unraveling the Links Underlying Arterial Stiffness, Bone Demineralization, and Muscle Loss. Hypertension 2020; 76:629-639. [DOI: 10.1161/hypertensionaha.120.15184] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effects of elevated arterial stiffness on cardiovascular outcomes are widely studied, whereas the relation to noncardiovascular outcomes relevant to older persons, such as the effect on bones and muscles, is less well established. Arterial stiffness, bone demineralization, and muscle loss are all age-related processes with common risk factors, however, whether these are just parallel age-related alterations or whether these processes share common pathways is not yet understood. In this review, we outline previous literature using different assessments of arterial stiffness in various populations across the world to produce a comprehensive overview. Although there are many studies showing an association between arterial stiffness and loss of bone and muscle, the majority are cross-sectional and there is limited longitudinal evidence to justify causal conclusions. We also give an in-depth review of hypotheses and possible mechanisms which may underlie these associations including hormone dysregulation, impaired glucose metabolism, and inflammation. This narrative review highlights the associations between vessels, bones, and muscles with aging, offering insights into possible shared pathways.
Collapse
Affiliation(s)
- Lisanne Tap
- From the Department of Internal Medicine, Section of Geriatric Medicine, Erasmus MC University Medical Center Rotterdam, The Netherlands (L.T., F.M.-R.)
| | - Frances A. Kirkham
- Brighton and Sussex Medical School, University of Sussex, United Kingdom (F.A.K., C.R.)
| | - Francesco Mattace-Raso
- From the Department of Internal Medicine, Section of Geriatric Medicine, Erasmus MC University Medical Center Rotterdam, The Netherlands (L.T., F.M.-R.)
| | - Laure Joly
- Department of Geriatrics, FHU-CARTAGE, CHRU de Nancy, and INSERM DCAC, Université de Lorraine, France (L.J., A.B.)
| | - Chakravarthi Rajkumar
- Brighton and Sussex Medical School, University of Sussex, United Kingdom (F.A.K., C.R.)
| | - Athanase Benetos
- Department of Geriatrics, FHU-CARTAGE, CHRU de Nancy, and INSERM DCAC, Université de Lorraine, France (L.J., A.B.)
| |
Collapse
|
37
|
Carrai P, Camarri S, Pondrelli CR, Gonnelli S, Caffarelli C. Calcification of Cardiac Valves in Metabolic Bone Disease: An Updated Review of Clinical Studies. Clin Interv Aging 2020; 15:1085-1095. [PMID: 32764895 PMCID: PMC7367930 DOI: 10.2147/cia.s244063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
Epidemiological and clinical data have suggested the existence of a relationship between cardiovascular diseases and metabolic bone disease. Several studies have demonstrated that heart valve calcification presents substantial similarities with that of bone. Literature data indicate that there are many active processes which promote osteogenesis and loss of mineralization inhibitors that lead to the deposition of extracellular matrix and proteins of bone tissue in cardiac valves. This review aimed to synthesize the available data in order to allow a better understanding of the relationship between osteoporosis or other metabolic bone diseases, such as primary hyperparathyroidism, and valvular calcification in humans. Electronic databases of Pubmed-Medline, Cochrane Library, and SCOPUS from inception to March 31, 2019 were searched. The full set of the articles potentially eligible were carefully assessed and reviewed. Finally, 23 studies were eligible and included in the systematic review. The majority of studies reported that osteoporosis and/or osteopenia were independent risk factors for valvular calcifications, even after adjusting for common cardiovascular risk factors. This suggests that this relationship is not only due to the presence of common cardiovascular risk factors but rather to underlying biological factors that connect them. Instead, regarding the association between primary hyperparathyroidism and valve calcification, conflicting data were found in the literature. To sum up, most of the literature data confirm that cardiac valve calcification processes are strongly influenced by alterations in bone metabolism. In particular, the patients with osteoporosis or primary hyperparathyroidism have an acceleration in the process of valvular calcification. Additional studies are needed to specifically address the mechanisms by which metabolic bone diseases could influence cardiac valve calcification.
Collapse
Affiliation(s)
- Paolo Carrai
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Silvia Camarri
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | | - Stefano Gonnelli
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Carla Caffarelli
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
38
|
Evenepoel P, Dejongh S, Verbeke K, Meijers B. The Role of Gut Dysbiosis in the Bone-Vascular Axis in Chronic Kidney Disease. Toxins (Basel) 2020; 12:toxins12050285. [PMID: 32365480 PMCID: PMC7290823 DOI: 10.3390/toxins12050285] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at increased risk of bone mineral density loss and vascular calcification. Bone demineralization and vascular mineralization often concur in CKD, similar to what observed in the general population. This contradictory association is commonly referred to as the 'calcification paradox' or the bone-vascular axis. Mounting evidence indicates that CKD-associated gut dysbiosis may be involved in the pathogenesis of the bone-vascular axis. A disrupted intestinal barrier function, a metabolic shift from a predominant saccharolytic to a proteolytic fermentation pattern, and a decreased generation of vitamin K may, alone or in concert, drive a vascular and skeletal pathobiology in CKD patients. A better understanding of the role of gut dysbiosis in the bone-vascular axis may open avenues for novel therapeutics, including nutriceuticals.
Collapse
Affiliation(s)
- Pieter Evenepoel
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven—University of Leuven, B-3000 Leuven, Belgium
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, B-3000 Leuven, Belgium
- Correspondence: ; Tel.: +32-16-344591; Fax: +32-16-344599
| | - Sander Dejongh
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven—University of Leuven, B-3000 Leuven, Belgium
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, B-3000 Leuven, Belgium
| | - Kristin Verbeke
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven—University of Leuven, B-3000 Leuven, Belgium
| | - Bjorn Meijers
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven—University of Leuven, B-3000 Leuven, Belgium
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
39
|
Asporin Reduces Adult Aortic Valve Interstitial Cell Mineralization Induced by Osteogenic Media and Wnt Signaling Manipulation In Vitro. Int J Cell Biol 2020; 2020:2045969. [PMID: 32328102 PMCID: PMC7171660 DOI: 10.1155/2020/2045969] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/30/2020] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Worldwide, calcific aortic valve disease is one of the leading causes of morbidity and mortality among patients with cardiac abnormalities. Aortic valve mineralization and calcification are the key events of adult calcific aortic valve disease manifestation and functional insufficiency. Due to heavy mineralization and calcification, adult aortic valvular cusps show disorganized and dispersed stratification concomitant with deposition of calcific nodules with severely compromised adult valve function. Interestingly, shared gene regulatory pathways are identified between bone-forming cells and heart valve cells during development. Asporin, a small leucine-rich proteoglycan (43 kDa), acts to inhibit mineralization in periodontal ligament cells and is also detected in normal murine adult aortic valve leaflets with unknown function. Therefore, to understand the Asporin function in aortic cusp mineralization and calcification, adult avian aortic valvular interstitial cell culture system is established and osteogenesis has been induced in these cells successfully. Upon induction of osteogenesis, reduced expression of Asporin mRNA and increased expression of bone and osteogenesis markers are detected compared to cells maintained without osteogenic induction. Importantly, treatment with human recombinant Asporin protein reduces the mineralization level in osteogenic media-induced aortic valvular interstitial cells with the concomitant decreased level of Wnt/β-catenin signaling. Overall, all these data are highly indicative that Asporin might be a novel biomolecular target to treat patients of calcific aortic valve disease over current cusp replacement surgery.
Collapse
|
40
|
Passos LSA, Lupieri A, Becker-Greene D, Aikawa E. Innate and adaptive immunity in cardiovascular calcification. Atherosclerosis 2020; 306:59-67. [PMID: 32222287 DOI: 10.1016/j.atherosclerosis.2020.02.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/10/2020] [Accepted: 02/20/2020] [Indexed: 12/19/2022]
Abstract
Despite the focus placed on cardiovascular research, the prevalence of vascular and valvular calcification is increasing and remains a leading contributor of cardiovascular morbidity and mortality. Accumulating studies provide evidence that cardiovascular calcification is an inflammatory disease in which innate immune signaling becomes sustained and/or excessive, shaping a deleterious adaptive response. The triggering immune factors and subsequent inflammatory events surrounding cardiovascular calcification remain poorly understood, despite sustained significant research interest and support in the field. Most studies on cardiovascular calcification focus on innate cells, particularly macrophages' ability to release pro-osteogenic cytokines and calcification-prone extracellular vesicles and apoptotic bodies. Even though substantial evidence demonstrates that macrophages are key components in triggering cardiovascular calcification, the crosstalk between innate and adaptive immune cell components has not been adequately addressed. The only therapeutic options currently used are invasive procedures by surgery or transcatheter intervention. However, no approved drug has shown prophylactic or therapeutic effectiveness. Conventional diagnostic imaging is currently the best method for detecting, measuring, and assisting in the treatment of calcification. However, these common imaging modalities are unable to detect early subclinical stages of disease at the level of microcalcifications; therefore, the vast majority of patients are diagnosed when macrocalcifications are already established. In this review, we unravel the current knowledge of how innate and adaptive immunity regulate cardiovascular calcification; and put forward differences and similarities between vascular and valvular disease. Additionally, we highlight potential immunomodulatory drugs with the potential to target calcification and propose avenues in need of further translational inquiry.
Collapse
Affiliation(s)
- Livia S A Passos
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Adrien Lupieri
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dakota Becker-Greene
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Pathology, Sechenov First Moscow State Medical University, Moscow, 119992, Russia.
| |
Collapse
|
41
|
Zhou P, Li Q, Su S, Dong W, Zong S, Ma Q, Yang X, Zuo D, Zheng S, Meng X, Xu D, Zeng Q. Interleukin 37 Suppresses M1 Macrophage Polarization Through Inhibition of the Notch1 and Nuclear Factor Kappa B Pathways. Front Cell Dev Biol 2020; 8:56. [PMID: 32117982 PMCID: PMC7033589 DOI: 10.3389/fcell.2020.00056] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022] Open
Abstract
Macrophage-orchestrated chronic inflammation plays an important role in cardiovascular disease, including accelerating the development of calcific aortic valve disease (CAVD). M1 and M2 macrophage polarization imbalances can alter intensity of inflammatory responses. Recombinant human interleukin 37 (IL-37) could be involved in regulating immune cell function to attenuate inflammation. This study aimed to identify IL-37 specifically modulates M1 polarization and investigate the underlying mechanism. Compared with normal valves, there are more M1 macrophages accumulation and less IL-37 expression in calcific aortic valves, which may indicate a negative relationship between IL-37 and M1 polarization. THP-1 cells could differentiate into resting macrophages with phorbol-12-myristate-13-acetate (PMA) and then polarize into M1 macrophages following treatment with lipopolysaccharide (LPS) and interferon gamma (IFN-γ). In vitro, recombinant human IL-37 attenuated the expression of inducible nitric oxide synthase (iNOS), CD11c, IL-6 and monocyte chemoattractant protein 1 (MCP-1) in M1 but augmented the expression of CD206 and IL-10 in M2. The suppression of M1 polarization was associated with the inhibition of the activation of the nuclear factor kappa B (NF-κB) and Notch1 signaling pathways. These results demonstrated that IL-37 inhibits the macrophages polarizing into M1 type via the inhibition of the Notch1 and nuclear factor kappa B pathways. In summary, IL-37 could be a potential therapeutic candidate for progressive CAVD by modulating M1 polarization and its orchestrated inflammation.
Collapse
Affiliation(s)
- Peitao Zhou
- Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianqin Li
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuwen Su
- Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenhui Dong
- Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyu Zong
- Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiong Ma
- Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Yang
- Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Daming Zuo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianzhong Meng
- Department of Surgery, University of Colorado Denver, Aurora, CO, United States
| | - Dingli Xu
- Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Qingchun Zeng
- Key Laboratory for Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Surgery, University of Colorado Denver, Aurora, CO, United States.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
42
|
de Oliveira Sá MPB, Cavalcanti LRP, Perazzo ÁM, Gomes RAF, Clavel MA, Pibarot P, Biondi-Zoccai G, Zhigalov K, Weymann A, Ruhparwar A, Lima RC. Calcific Aortic Valve Stenosis and Atherosclerotic Calcification. Curr Atheroscler Rep 2020; 22:2. [PMID: 31912380 DOI: 10.1007/s11883-020-0821-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review summarizes the pathophysiology of calcific aortic valve stenosis (CAVS) and surveys relevant clinical data and basic research that explain how CAVS arises. RECENT FINDINGS Lipoprotein(a) [Lp(a)], lipoprotein-associated phospholipase A2 (Lp-PLA2), oxidized phospholipids (OxPL), autotaxin, and genetic driving forces such as mutations in LPA gene and NOTCH gene seem to play a major role in the development of CAVS. These factors might well become targets of medical therapy in the coming years. CVAS seems to be a multifactorial disease that has much in common with coronary artery disease, mainly regarding lipidic accumulation and calcium deposition. No clinical trials conducted to date have managed to answer the key question of whether Lp(a) lowering and anti-calcific therapies confer a benefit in terms of reducing incidence or progression of CAVS, although additional outcome trials are ongoing.
Collapse
Affiliation(s)
- Michel Pompeu Barros de Oliveira Sá
- Division of Cardiovascular Surgery of Pronto Socorro Cardiológico de Pernambuco, PROCAPE, Av. Eng. Domingos Ferreira n°4172. Edf. Paquetá, apt° 405, Recife, Pernambuco, 51021-040, Brazil.
- University of Pernambuco, UPE, Recife, Brazil.
- Nucleus of Postgraduate and Research in Health Sciences of Faculty of Medical Sciences and Biological Sciences Instituite, FCM/ICB, Recife, Brazil.
| | - Luiz Rafael P Cavalcanti
- Division of Cardiovascular Surgery of Pronto Socorro Cardiológico de Pernambuco, PROCAPE, Av. Eng. Domingos Ferreira n°4172. Edf. Paquetá, apt° 405, Recife, Pernambuco, 51021-040, Brazil
- University of Pernambuco, UPE, Recife, Brazil
| | - Álvaro M Perazzo
- Division of Cardiovascular Surgery of Pronto Socorro Cardiológico de Pernambuco, PROCAPE, Av. Eng. Domingos Ferreira n°4172. Edf. Paquetá, apt° 405, Recife, Pernambuco, 51021-040, Brazil
- University of Pernambuco, UPE, Recife, Brazil
| | - Rafael A F Gomes
- Division of Cardiovascular Surgery of Pronto Socorro Cardiológico de Pernambuco, PROCAPE, Av. Eng. Domingos Ferreira n°4172. Edf. Paquetá, apt° 405, Recife, Pernambuco, 51021-040, Brazil
- University of Pernambuco, UPE, Recife, Brazil
- Nucleus of Postgraduate and Research in Health Sciences of Faculty of Medical Sciences and Biological Sciences Instituite, FCM/ICB, Recife, Brazil
| | - Marie-Annick Clavel
- Québec Heart and Lung Institute/Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, 2725 Chemin Sainte Foy, #A-2075, Quebec, QC, G1V4G5, Canada
| | - Philippe Pibarot
- Québec Heart and Lung Institute/Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, 2725 Chemin Sainte Foy, #A-2075, Quebec, QC, G1V4G5, Canada
| | - Giuseppe Biondi-Zoccai
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Konstantin Zhigalov
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, University Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Alexander Weymann
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, University Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Arjang Ruhparwar
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, University Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Ricardo Carvalho Lima
- Division of Cardiovascular Surgery of Pronto Socorro Cardiológico de Pernambuco, PROCAPE, Av. Eng. Domingos Ferreira n°4172. Edf. Paquetá, apt° 405, Recife, Pernambuco, 51021-040, Brazil
- University of Pernambuco, UPE, Recife, Brazil
- Nucleus of Postgraduate and Research in Health Sciences of Faculty of Medical Sciences and Biological Sciences Instituite, FCM/ICB, Recife, Brazil
| |
Collapse
|
43
|
Gee T, Farrar E, Wang Y, Wu B, Hsu K, Zhou B, Butcher J. NFκB (Nuclear Factor κ-Light-Chain Enhancer of Activated B Cells) Activity Regulates Cell-Type-Specific and Context-Specific Susceptibility to Calcification in the Aortic Valve. Arterioscler Thromb Vasc Biol 2020; 40:638-655. [PMID: 31893948 DOI: 10.1161/atvbaha.119.313248] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Although often studied independently, little is known about how aortic valve endothelial cells and valve interstitial cells interact collaborate to maintain tissue homeostasis or drive valve calcific pathogenesis. Inflammatory signaling is a recognized initiator of valve calcification, but the cell-type-specific downstream mechanisms have not been elucidated. In this study, we test how inflammatory signaling via NFκB (nuclear factor κ-light-chain enhancer of activated B cells) activity coordinates unique and shared mechanisms of valve endothelial cells and valve interstitial cells differentiation during calcific progression. Approach and Results: Activated NFκB was present throughout the calcific aortic valve disease (CAVD) process in both endothelial and interstitial cell populations in an established mouse model of hypercholesterolemia-induced CAVD and in human CAVD. NFκB activity induces endothelial to mesenchymal transformation in 3-dimensional cultured aortic valve endothelial cells and subsequent osteogenic calcification of transformed cells. Similarly, 3-dimensional cultured valve interstitial cells calcified via NFκB-mediated osteogenic differentiation. NFκB-mediated endothelial to mesenchymal transformation was directly demonstrated in vivo during CAVD via genetic lineage tracking. Genetic deletion of NFκB in either whole valves or valve endothelium only was sufficient to prevent valve-specific molecular and cellular mechanisms of CAVD in vivo despite the persistence of a CAVD inducing environment. CONCLUSIONS Our results identify NFκB signaling as an essential molecular regulator for both valve endothelial and interstitial participation in CAVD pathogenesis. Direct demonstration of valve endothelial cell endothelial to mesenchymal transformation transmigration in vivo during CAVD highlights a new cellular population for further investigation in CAVD morbidity. The efficacy of valve-specific NFκB modulation in inhibiting hypercholesterolemic CAVD suggests potential benefits of multicell type integrated investigation for biological therapeutic development and evaluation for CAVD.
Collapse
Affiliation(s)
- Terence Gee
- From the Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY (T.G., E.F., K.H., J.B.)
| | - Emily Farrar
- From the Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY (T.G., E.F., K.H., J.B.)
| | - Yidong Wang
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (Y.W., B.W., B.Z.)
| | - Bingruo Wu
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (Y.W., B.W., B.Z.)
| | - Kevin Hsu
- From the Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY (T.G., E.F., K.H., J.B.)
| | - Bin Zhou
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (Y.W., B.W., B.Z.)
| | - Jonathan Butcher
- From the Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY (T.G., E.F., K.H., J.B.)
| |
Collapse
|
44
|
|
45
|
Rogers MA, Aikawa E. MicroRNA Extracellular Vesicle Stowaways in Cell-Cell Communication and Organ Crosstalk. Arterioscler Thromb Vasc Biol 2019; 39:2448-2450. [PMID: 31770027 DOI: 10.1161/atvbaha.119.313533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Maximillian A Rogers
- From the Center for Interdisciplinary Cardiovascular Sciences (M.A.R., E.A.), Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- From the Center for Interdisciplinary Cardiovascular Sciences (M.A.R., E.A.), Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Center for Excellence in Vascular Biology (E.A.), Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Pathology, Sechenov First Moscow State Medical University Moscow, Russia (E.A.)
| |
Collapse
|
46
|
Lipoprotein(a) as Orchestrator of Calcific Aortic Valve Stenosis. Biomolecules 2019; 9:biom9120760. [PMID: 31766423 PMCID: PMC6995555 DOI: 10.3390/biom9120760] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022] Open
Abstract
Aortic valve stenosis (AVS) is the most prevalent valvular heart disease in the Western World with exponentially increased incidence with age. If left untreated, the yearly mortality rates increase up to 25%. Currently, no effective pharmacological interventions have been established to treat or prevent AVS. The only treatment modality so far is surgical or transcatheter aortic valve replacement (AVR). Lipoprotein(a) [Lp(a)] has been implicated as a pivotal player in the pathophysiology of calcification of the valves. Patients with elevated levels of Lp(a) have a higher risk of hospitalization or mortality due to the presence of AVS. Multiple studies indicated Lp(a) as a likely causal and independent risk factor for AVS. This review discusses the most important findings and mechanisms related to Lp(a) and AVS in detail. During the progression of AVS, Lp(a) enters the aortic valve tissue at damaged sites of the valves. Subsequently, autotaxin converts lysophosphatidylcholine in lysophosphatidic acid (LysoPA) which in turn acts as a ligand for the LysoPA receptor. This triggers a nuclear factor-κB cascade leading to increased transcripts of interleukin 6, bone morphogenetic protein 2, and runt-related transcription factor 2. This progresses to the actual calcification of the valves through production of alkaline phosphatase and calcium depositions. Furthermore, this review briefly mentions potentially interesting therapies that may play a role in the treatment or prevention of AVS in the near future.
Collapse
|
47
|
Evenepoel P, Opdebeeck B, David K, D'Haese PC. Bone-Vascular Axis in Chronic Kidney Disease. Adv Chronic Kidney Dis 2019; 26:472-483. [PMID: 31831125 DOI: 10.1053/j.ackd.2019.09.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/30/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022]
Abstract
Patients with chronic kidney disease (CKD) are at increased risk of osteoporosis and vascular calcification. Bone demineralization and vascular mineralization go often hand in hand in CKD, similar to as in the general population. This contradictory association is independent of aging and is commonly referred to as the "calcification paradox" or the bone-vascular axis. Various common risk factors and mechanisms have been identified. Alternatively, calcifying vessels may release circulating factors that affect bone metabolism, while bone disease may infer conditions that favor vascular calcification. The present review focuses on emerging concepts and major mechanisms involved in the bone-vascular axis in the setting of CKD. A better understanding of these concepts and mechanisms may identify therapeutics able to target and exert beneficial effects on bone and vasculature simultaneously.
Collapse
|
48
|
Creager MD, Hohl T, Hutcheson JD, Moss AJ, Schlotter F, Blaser MC, Park MA, Lee LH, Singh SA, Alcaide-Corral CJ, Tavares AAS, Newby DE, Kijewski MF, Aikawa M, Di Carli M, Dweck MR, Aikawa E. 18F-Fluoride Signal Amplification Identifies Microcalcifications Associated With Atherosclerotic Plaque Instability in Positron Emission Tomography/Computed Tomography Images. Circ Cardiovasc Imaging 2019; 12:e007835. [PMID: 30642216 DOI: 10.1161/circimaging.118.007835] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Microcalcifications in atherosclerotic plaques are destabilizing, predict adverse cardiovascular events, and are associated with increased morbidity and mortality.18F-fluoride positron emission tomography (PET)/computed tomography (CT) imaging has demonstrated promise as a useful clinical diagnostic tool in identifying high-risk plaques; however, there is confusion as to the underlying mechanism of signal amplification seen in PET-positive, CT-negative image regions. This study tested the hypothesis that 18F-fluoride PET/CT can identify early microcalcifications. METHODS 18F-fluoride signal amplification derived from microcalcifications was validated against near-infrared fluorescence molecular imaging and histology using an in vitro 3-dimensional hydrogel collagen platform, ex vivo human specimens, and a mouse model of atherosclerosis. RESULTS Microcalcification size correlated inversely with collagen concentration. The 18F-fluoride ligand bound to microcalcifications formed by calcifying vascular smooth muscle cell derived extracellular vesicles in the in vitro 3-dimensional collagen system and exhibited an increasing signal with an increase in collagen concentration (0.25 mg/mL collagen -33.8×102±12.4×102 counts per minute; 0.5 mg/mL collagen -67.7×102±37.4×102 counts per minute; P=0.0014), suggesting amplification of the PET signal by smaller microcalcifications. We further incubated human atherosclerotic endarterectomy specimens with clinically relevant concentrations of 18F-fluoride. The 18F-fluoride ligand labeled microcalcifications in PET-positive, CT-negative regions of explanted human specimens as evidenced by 18F-fluoride PET/CT imaging, near-infrared fluorescence, and histological analysis. Additionally, the 18F-fluoride ligand identified micro and macrocalcifications in atherosclerotic aortas obtained from low-density lipoprotein receptor-deficient mice. CONCLUSIONS Our results suggest that 18F-fluoride PET signal in PET-positive, CT-negative regions of human atherosclerotic plaques is the result of developing microcalcifications, and high surface area in regions of small microcalcifications may amplify PET signal.
Collapse
Affiliation(s)
- Michael D Creager
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Tobias Hohl
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | | | - Alastair J Moss
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - Florian Schlotter
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Mark C Blaser
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Mi-Ae Park
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.-A.P., M.F.K., M.D.C.)
| | - Lang Ho Lee
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Sasha A Singh
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Carlos J Alcaide-Corral
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - Adriana A S Tavares
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - David E Newby
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - Marie F Kijewski
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.-A.P., M.F.K., M.D.C.)
| | - Masanori Aikawa
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.).,Division of Cardiovascular Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.A., E.A.)
| | - Marcelo Di Carli
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.-A.P., M.F.K., M.D.C.)
| | - Marc R Dweck
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.).,Division of Cardiovascular Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.A., E.A.)
| |
Collapse
|
49
|
Teplyakov AT, Berezikova EN, Shilov SN, Popova AA, Samsonova EN, Yakovleva IV, Molokov AV, Grakova EV, Kopeva KV. [The effect of bisphosphonate therapy on reducing the risk of cardiovascular complications associated with chronic heart failure, type 2 diabetes and osteoporosis in postmenopausal women]. TERAPEVT ARKH 2019; 91:63-69. [PMID: 32598633 DOI: 10.26442/00403660.2019.10.000162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Indexed: 11/22/2022]
Abstract
AIM To study the effectiveness of oral alendronate and ibandronate bisphosphonates for the prevention of cardiovascular complications in postmenopausal women with type 2 diabetes mellitus (DM) and osteoporosis during a 12-month prospective observation. MATERIALS AND METHODS The study included 86 women with osteoporosis, chronic heart failure (CHF) and type 2 diabetes: the 1st group (n=52) included patients who received basic therapy for heart failure; the 2nd group (n=34) included patients who, in addition to the basic therapy of heart failure, were prescribed alendronic and ibandronic acid preparations for the treatment of osteoporosis. In order to identify the possibility of associating the studied factors with the nature of the course of heart failure, the patients were divided according to the results of a one - year follow - up into two subgroups: subgroup A (n=49) - patients with a favorable course of the disease and subgroup B (n=37) - patients with an unfavorable course of pathology. RESULTS AND DISCUSSION After 12 months, a significant decrease in the levels of cerebral natriuretic peptide precursor (NT-proBNP), tumor necrosis factor-α, and interleukin-1β was found in the group of women treated with bisphosphonates compared to baseline. Significant associations of NT-proBNP levels (p=0.02) and the studied cytokines (p=0.01) with an unfavorable course of heart failure were revealed. A significant association of bisphosphonate therapy with a favorable course of heart failure (p=0.01) was also revealed. The probability of developing adverse cardiovascular events during the year in the treatment of heart failure with basic therapy drugs with additional therapy of osteoporosis with bisphosphonates is significantly (p=0.0025) lower than the treatment of patients with heart failure with only basic therapy and not taking bisphosphonates for the treatment of osteoporosis. CONCLUSION In postmenopausal women with associated cardiovascular pathology (CHF, type 2 diabetes and osteoporosis), prophylactic therapy with oral alendronate and ibandronate oral bisphosphonates is effective, reduces the risk of progression of heart failure, inhibits inflammatory mediators, positively affects the combined endpoints of comorbid cardiovascular pathology.
Collapse
Affiliation(s)
- A T Teplyakov
- Cardiology Research Institute, Tomsk National Research Medical Center
| | | | | | | | | | | | | | - E V Grakova
- Cardiology Research Institute, Tomsk National Research Medical Center
| | - K V Kopeva
- Cardiology Research Institute, Tomsk National Research Medical Center
| |
Collapse
|
50
|
Abstract
Vascular calcification (VC) is strongly associated with all-cause mortality and is an independent predictor of cardiovascular events. Resulting from its complex, multifaceted nature, targeted treatments for VC have not yet been developed. Lipoproteins are well characterized in the pathogenesis of atherosclerotic plaques, leading to the development of plaque regressing therapeutics. Although their roles in plaque progression are well documented, their roles in VC, and calcification of a plaque, are not well understood. In this review, early in vitro data and clinical correlations suggest an inhibitory role for HDL (high-density lipoproteins) in VC, a stimulatory role for LDL (low-density lipoprotein) and VLDL (very low-density lipoprotein) and a potentially causal role for Lp(a) (lipoprotein [a]). Additionally, after treatment with a statin or PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitor, plaque calcification is observed to increase. With the notion that differing morphologies of plaque calcification associate with either a more stable or unstable plaque phenotype, uncovering the mechanisms of lipoprotein-artery wall interactions could produce targeted therapeutic options for VC.
Collapse
Affiliation(s)
- Emma J. Akers
- From the South Australian Health and Medical Research Institute, Adelaide, Australia (E.J.A.)
- The University of Adelaide, Australia (E.J.A.)
| | - Stephen J. Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Australia (S.J.N.)
| | - Belinda A. Di Bartolo
- The Kolling Institute of Medical Research, The University of Sydney, Australia (B.A.D.B.)
| |
Collapse
|