151
|
Rogers MA, Buffolo F, Schlotter F, Atkins SK, Lee LH, Halu A, Blaser MC, Tsolaki E, Higashi H, Luther K, Daaboul G, Bouten CVC, Body SC, Singh SA, Bertazzo S, Libby P, Aikawa M, Aikawa E. Annexin A1-dependent tethering promotes extracellular vesicle aggregation revealed with single-extracellular vesicle analysis. SCIENCE ADVANCES 2020; 6:eabb1244. [PMID: 32938681 PMCID: PMC7494353 DOI: 10.1126/sciadv.abb1244] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 07/29/2020] [Indexed: 06/11/2023]
Abstract
Extracellular vesicles (EVs) including plasma membrane-derived microvesicles and endosomal-derived exosomes aggregate by unknown mechanisms, forming microcalcifications that promote cardiovascular disease, the leading cause of death worldwide. Here, we show a framework for assessing cell-independent EV mechanisms in disease by suggesting that annexin A1 (ANXA1)-dependent tethering induces EV aggregation and microcalcification. We present single-EV microarray, a method to distinguish microvesicles from exosomes and assess heterogeneity at a single-EV level. Single-EV microarray and proteomics revealed increased ANXA1 primarily on aggregating and calcifying microvesicles. ANXA1 vesicle aggregation was suppressed by calcium chelation, altering pH, or ANXA1 neutralizing antibody. ANXA1 knockdown attenuated EV aggregation and microcalcification formation in human cardiovascular cells and acellular three-dimensional collagen hydrogels. Our findings explain why microcalcifications are more prone to form in vulnerable regions of plaque, regulating critical cardiovascular pathology, and likely extend to other EV-associated diseases, including autoimmune and neurodegenerative diseases and cancer.
Collapse
Affiliation(s)
- Maximillian A Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Fabrizio Buffolo
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Florian Schlotter
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Samantha K Atkins
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lang H Lee
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Arda Halu
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Tsolaki
- Department of Medical Physics and Biomedical Engineering, University College London, London WC1E6BT, UK
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5612, Netherlands
| | - Simon C Body
- Department of Anesthesiology, Boston University School of Medicine, Boston, MA 02218, USA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering, University College London, London WC1E6BT, UK
| | - Peter Libby
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow 119992, Russia
| |
Collapse
|
152
|
Cheng CI, Chang HR, Tai MH, Chou MH, Chen GT, Chen PH, Kao YH. Hepatoma-derived growth factor enhances osteoblastic transformation of rat aortic vascular smooth muscle cells in vitro. Life Sci 2020; 256:117964. [DOI: 10.1016/j.lfs.2020.117964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023]
|
153
|
Karpouzas GA, Ormseth SR, Hernandez E, Budoff MJ. Biologics May Prevent Cardiovascular Events in Rheumatoid Arthritis by Inhibiting Coronary Plaque Formation and Stabilizing High‐Risk Lesions. Arthritis Rheumatol 2020; 72:1467-1475. [DOI: 10.1002/art.41293] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 04/16/2020] [Indexed: 12/14/2022]
Affiliation(s)
- George A. Karpouzas
- Harbor‐UCLA Medical Center and Lundquist Institute for Biomedical Innovation Torrance California
| | - Sarah R. Ormseth
- Harbor‐UCLA Medical Center and Lundquist Institute for Biomedical Innovation Torrance California
| | - Elizabeth Hernandez
- Harbor‐UCLA Medical Center and Lundquist Institute for Biomedical Innovation Torrance California
| | - Matthew J. Budoff
- Harbor‐UCLA Medical Center and Lundquist Institute for Biomedical Innovation Torrance California
| |
Collapse
|
154
|
Daghem M, Newby DE. Innovation in medical imaging to improve disease staging, therapeutic intervention, and clinical outcomes. Atherosclerosis 2020; 306:75-84. [DOI: 10.1016/j.atherosclerosis.2020.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/20/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022]
|
155
|
Ghosh S, He W, Gao J, Luo D, Wang J, Chen J, Huang H. Whole milk consumption is associated with lower risk of coronary artery calcification progression: evidences from the Multi-Ethnic Study of Atherosclerosis. Eur J Nutr 2020; 60:1049-1058. [PMID: 32583016 DOI: 10.1007/s00394-020-02301-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Coronary artery calcification (CAC) progression is a strong predictor of cardiovascular disease (CVD) morbidity and mortality. However, the association between whole milk and CAC progression remains unknown. Recent studies highlighted beneficial effects of short chain fatty acids (SCFA) from whole milk on CVD. In this study, we attempted to investigate the relationship between whole milk consumption and CAC progression, and the potential effect of SCFA in it. METHODS We analyzed a population-based cohort with 5273 participants from the Multi-Ethnic Study of Atherosclerosis (MESA) who completed a dietary questionnaire at baseline. CAC was measured at baseline and subsequent follow-up examinations by multi-detector computed tomography (MDCT) scans with Agatston scores. CAC progression was defined as increased CAC scores in the follow-up from the baseline exam. RESULTS Participants consuming whole milk exhibited lower baseline CAC and CAC progression than those who never/rarely consumed whole milk (P < 0.001 and P = 0.010, respectively). Moreover, multivariable logistic regression analysis demonstrated that whole milk intake was independently associated with lower CAC progression (OR 0.765; 95% CI 0.600-0.977; P = 0.032), especially in males, participants with age ≤ 64 years and with body mass index (BMI) ≤ 25 kg/m2. Mediation analysis further showed that caproic acid, one kind of SCFA, partly mediated protective effects of whole milk on CAC progression. CONCLUSIONS Self-reported whole milk consumption was inversely associated with CAC progression in community-dwelling participants, especially in those at relatively low cardiovascular risks. The beneficial effect was partially mediated by SCFA. Therefore, whole milk can be incorporated into part of a cardio-protective diet. Regarding this, future studies may target SCFA to provide insight into more mechanistic views.
Collapse
Affiliation(s)
- Sounak Ghosh
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Wanbing He
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jingwei Gao
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dongling Luo
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jingfeng Wang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Chen
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Hui Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
156
|
Li C, Zhang S, Chen X, Ji J, Yang W, Gui T, Gai Z, Li Y. Farnesoid X receptor activation inhibits TGFBR1/TAK1-mediated vascular inflammation and calcification via miR-135a-5p. Commun Biol 2020; 3:327. [PMID: 32581266 PMCID: PMC7314757 DOI: 10.1038/s42003-020-1058-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 06/08/2020] [Indexed: 01/06/2023] Open
Abstract
Chronic inflammation plays a crucial role in vascular calcification. However, only a few studies have revealed the mechanisms underlying the development of inflammation under high-phosphate conditions in chronic kidney disease (CKD) patients. Here, we show that inflammation resulting from the activation of the TGFBR1/TAK1 pathway is involved in calcification in CKD rats or osteogenic medium-cultured human aortic smooth muscle cells (HASMCs). Moreover, miR-135a-5p is demonstrated to be a key regulator of the TGFBR1/TAK1 pathway, which has been reported to be decreased in CKD rats. We further reveal that farnesoid X receptor (FXR) activation increases miR-135a-5p expression, thereby inhibiting the activation of the TGFBR1/TAK1 pathway, ultimately resulting in the attenuation of vascular inflammation and calcification in CKD rats. Our findings provide advanced insights into the mechanisms underlying the development of inflammation in vascular calcification, and evidence that FXR activation could serve as a therapeutic strategy for retarding vascular calcification in CKD patients.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Calcinosis/genetics
- Calcinosis/metabolism
- Cells, Cultured
- Female
- Humans
- MAP Kinase Kinase Kinases/genetics
- MAP Kinase Kinase Kinases/metabolism
- Male
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/cytology
- Osteogenesis
- Rats, Wistar
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/pathology
- Vasculitis/genetics
- Vasculitis/metabolism
- Vasculitis/pathology
Collapse
Affiliation(s)
- Chao Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, 8032, Switzerland
| | - Shijun Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaoqing Chen
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jingkang Ji
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenqing Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ting Gui
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhibo Gai
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, 8032, Switzerland.
| | - Yunlun Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, China.
| |
Collapse
|
157
|
Kawakami R, Katsuki S, Travers R, Romero DC, Becker-Greene D, Passos LSA, Higashi H, Blaser MC, Sukhova GK, Buttigieg J, Kopriva D, Schmidt AM, Anderson DG, Singh SA, Cardoso L, Weinbaum S, Libby P, Aikawa M, Croce K, Aikawa E. S100A9-RAGE Axis Accelerates Formation of Macrophage-Mediated Extracellular Vesicle Microcalcification in Diabetes Mellitus. Arterioscler Thromb Vasc Biol 2020; 40:1838-1853. [PMID: 32460581 DOI: 10.1161/atvbaha.118.314087] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Vascular calcification is a cardiovascular risk factor and accelerated in diabetes mellitus. Previous work has established a role for calcification-prone extracellular vesicles in promoting vascular calcification. However, the mechanisms by which diabetes mellitus provokes cardiovascular events remain incompletely understood. Our goal was to identify that increased S100A9 promotes the release of calcification-prone extracellular vesicles from human macrophages in diabetes mellitus. Approach and Results: Human primary macrophages exposed to high glucose (25 mmol/L) increased S100A9 secretion and the expression of receptor for advanced glycation end products (RAGE) protein. Recombinant S100A9 induced the expression of proinflammatory and osteogenic factors, as well as the number of extracellular vesicles with high calcific potential (alkaline phosphatase activity, P<0.001) in macrophages. Treatment with a RAGE antagonist or silencing with S100A9 siRNA in macrophages abolished these responses, suggesting that stimulation of the S100A9-RAGE axis by hyperglycemia favors a procalcific environment. We further showed that an imbalance between Nrf-2 (nuclear factor 2 erythroid related factor 2) and NF-κB (nuclear factor-κB) pathways contributes to macrophage activation and promotes a procalcific environment. In addition, streptozotocin-induced diabetic Apoe-/-S100a9-/- mice and mice treated with S100a9 siRNA encapsulated in macrophage-targeted lipid nanoparticles showed decreased inflammation and microcalcification in atherosclerotic plaques, as gauged by molecular imaging and comprehensive histological analysis. In human carotid plaques, comparative proteomics in patients with diabetes mellitus and histological analysis showed that the S100A9-RAGE axis associates with osteogenic activity and the formation of microcalcification. CONCLUSIONS Under hyperglycemic conditions, macrophages release calcific extracellular vesicles through mechanisms involving the S100A9-RAGE axis, thus contributing to the formation of microcalcification within atherosclerotic plaques.
Collapse
Affiliation(s)
- Ryo Kawakami
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shunsuke Katsuki
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Richard Travers
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dayanna Carolina Romero
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dakota Becker-Greene
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Livia Silva Araujo Passos
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Galina K Sukhova
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Josef Buttigieg
- Department of Biology, University of Regina, Saskatchewan, Canada (J.B.)
| | - David Kopriva
- Regina Qu'Appelle Health Region, University of Saskatchewan, Regina, Canada (D.K.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University (A.M.S.)
| | - Daniel G Anderson
- Institutes for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge (D.G.A.)
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Luis Cardoso
- Department of Biomedical Engineering, The City College of New York (L.C., S.W.)
| | - Sheldon Weinbaum
- Department of Biomedical Engineering, The City College of New York (L.C., S.W.)
| | - Peter Libby
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kevin Croce
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Human Pathology, Sechenov First Moscow State Medical University, Russia (E.A.)
| |
Collapse
|
158
|
Abstract
PURPOSE OF REVIEW This review summarizes the effects of microparticles and exosomes in the progression of atherosclerosis and the prospect for their diagnostic and therapeutic potentials. RECENT FINDINGS Microparticles and exosomes can induce endothelial dysfunction, vascular inflammation, coagulation, thrombosis, and calcification via their components of proteins and noncoding RNAs, which may promote the progression of atherosclerosis. The applications of microparticles and exosomes become the spotlight of clinical diagnosis and therapy. Microparticles and exosomes are members of extracellular vesicles, which are generated in various cell types by different mechanisms of cell membrane budding and multivesicular body secretion, respectively. They are important physiologic pathways of cell-to-cell communication in vivo and act as messengers accelerating or alleviating the process of atherosclerosis. Microparticles and exosomes may become diagnostic biomarkers and therapeutic approaches of atherosclerosis.
Collapse
|
159
|
Burgmaier M, Milzi A, Dettori R, Burgmaier K, Hellmich M, Almalla M, Marx N, Reith S. Colocalization of plaque macrophages and calcification in coronary plaques as detected by optical coherence tomography predicts cardiovascular outcome. Cardiol J 2020; 27:303-306. [PMID: 32436586 DOI: 10.5603/cj.a2020.0075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 01/25/2023] Open
Affiliation(s)
- Mathias Burgmaier
- Department of Cardiology, University Hospital of the RWTH Aachen, Pauwelstr. 30, 52062, Germany
| | - Andrea Milzi
- Department of Cardiology, University Hospital of the RWTH Aachen, Pauwelstr. 30, 52062, Germany
| | - Rosalia Dettori
- Department of Cardiology, University Hospital of the RWTH Aachen, Pauwelstr. 30, 52062, Germany
| | - Kathrin Burgmaier
- Department of Pediatrics, University Hospital of Cologne, Kerpener Str. 62, 50937, Germany
| | - Martin Hellmich
- Institute of Medical Statistics and Computational Biology (IMSB), University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Mohammad Almalla
- Department of Cardiology, University Hospital of the RWTH Aachen, Pauwelstr. 30, 52062, Germany
| | - Nikolaus Marx
- Department of Cardiology, University Hospital of the RWTH Aachen, Pauwelstr. 30, 52062, Germany
| | - Sebastian Reith
- Department of Cardiology, University Hospital of the RWTH Aachen, Pauwelstr. 30, 52062, Germany.
| |
Collapse
|
160
|
Tanoren B, Ugurlucan M, Ates Alkan F. Analysis of atherosclerosis by inductively coupled plasma optical emission spectroscopy and scanning acoustic microscopy. Biomed Phys Eng Express 2020; 6:045004. [DOI: 10.1088/2057-1976/ab8e96] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
161
|
Hou YC, Lu CL, Zheng CM, Liu WC, Yen TH, Chen RM, Lin YF, Chao CT, Lu KC. The Role of Vitamin D in Modulating Mesenchymal Stem Cells and Endothelial Progenitor Cells for Vascular Calcification. Int J Mol Sci 2020; 21:2466. [PMID: 32252330 PMCID: PMC7177675 DOI: 10.3390/ijms21072466] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Vascular calcification, which involves the deposition of calcifying particles within the arterial wall, is mediated by atherosclerosis, vascular smooth muscle cell osteoblastic changes, adventitial mesenchymal stem cell osteoblastic differentiation, and insufficiency of the calcification inhibitors. Recent observations implied a role for mesenchymal stem cells and endothelial progenitor cells in vascular calcification. Mesenchymal stem cells reside in the bone marrow and the adventitial layer of arteries. Endothelial progenitor cells that originate from the bone marrow are an important mechanism for repairing injured endothelial cells. Mesenchymal stem cells may differentiate osteogenically by inflammation or by specific stimuli, which can activate calcification. However, the bioactive substances secreted from mesenchymal stem cells have been shown to mitigate vascular calcification by suppressing inflammation, bone morphogenetic protein 2, and the Wingless-INT signal. Vitamin D deficiency may contribute to vascular calcification. Vitamin D supplement has been used to modulate the osteoblastic differentiation of mesenchymal stem cells and to lessen vascular injury by stimulating adhesion and migration of endothelial progenitor cells. This narrative review clarifies the role of mesenchymal stem cells and the possible role of vitamin D in the mechanisms of vascular calcification.
Collapse
Affiliation(s)
- Yi-Chou Hou
- Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, New Taipei City 231, Taiwan;
- School of Medicine, Fu-Jen Catholic University, New Taipei City 234, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-M.Z.); (W.-C.L.); (Y.-F.L.)
| | - Chien-Lin Lu
- School of Medicine, Fu-Jen Catholic University, New Taipei City 234, Taiwan;
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City 243, Taiwan
| | - Cai-Mei Zheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-M.Z.); (W.-C.L.); (Y.-F.L.)
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan
| | - Wen-Chih Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-M.Z.); (W.-C.L.); (Y.-F.L.)
- Division of Nephrology, Department of Internal Medicine, Tungs’ Taichung Metroharbor Hospital, Taichung City 43304, Taiwan
| | - Tzung-Hai Yen
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ruei-Ming Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-M.Z.); (W.-C.L.); (Y.-F.L.)
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan
| | - Chia-Ter Chao
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 104, Taiwan
- Nephrology division, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 108, Taiwan
| | - Kuo-Cheng Lu
- School of Medicine, Fu-Jen Catholic University, New Taipei City 234, Taiwan;
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City 243, Taiwan
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
162
|
Huang S, Yu X, Wang H, Zheng J. Elevated serum sortilin is related to carotid plaque concomitant with calcification. Biomark Med 2020; 14:381-389. [PMID: 32077308 DOI: 10.2217/bmm-2019-0472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: To explore whether elevated serum sortilin was associated with calcified carotid plaque and ischemic stroke. Methods: A total of 171 patients with cardiovascular risk factors were enrolled. Ultrasonography was performed to evaluate calcified plaques and noncalcified plaques. Serum sortilin concentration was measured by ELISA. Results: Serum sortilin level was higher in patients with calcified carotid plaque and positively related to carotid plaque burden, but not with ischemic stroke during the follow-up. Multivariable logistic regression analysis revealed serum sortilin level was an independent determinant for calcified carotid plaque (p = 0.001). Receiving operating characteristic analysis showed an area under the curve of sortilin for carotid calcification was 0.759. Conclusion: Higher serum sortilin level was associated with carotid calcification and severe carotid plaque score.
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Ultrasound, Shenzhen Second People’s Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, Guangdong, PR China
| | - Xingxing Yu
- Department of Internal Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang, PR China
| | - Haiqing Wang
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang, PR China
| | - Jianlei Zheng
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang, PR China
| |
Collapse
|
163
|
Li Z, Zhao Z, Cai Z, Sun Y, Li L, Yao F, Yang L, Zhou Y, Zhu H, Fu Y, Wang L, Fang W, Chen Y, Kong W. Runx2 (Runt-Related Transcription Factor 2)-Mediated Microcalcification Is a Novel Pathological Characteristic and Potential Mediator of Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2020; 40:1352-1369. [PMID: 32212850 DOI: 10.1161/atvbaha.119.314113] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Abdominal aortic aneurysms (AAAs) are highly lethal diseases without effective clinical predictors and therapeutic targets. Vascular microcalcification, as detected by fluorine-18-sodium fluoride, has recently been recognized as a valuable indicator in predicting atherosclerotic plaque rupture and AAA expansion. However, whether vascular microcalcification involved in the pathogenesis of AAA remains elusive. Approach and Results: Microcalcification was analyzed in human aneurysmal aortas histologically and in AngII (angiotensin II)-infused ApoE-/- mouse aortas by fluorine-18-sodium fluoride positron emission tomography and X-ray computed tomography scanning in chronological order in live animals. AAA patients' aortic tissue showed markedly enhanced microcalcification in the aortic media within the area proximal to elastic fiber degradation, compared with non-AAA patients. Enhanced fluorine-18-sodium fluoride uptake preceded significant aortic expansion in mice. Microcalcification-positive mice on day 7 of AngII infusion showed dramatic aortic expansion on subsequent days 14 to 28, whereas microcalcification-negative AngII-infused mice and saline-induced mice did not develop AAA. The application of hydroxyapatite, the main component of microcalcification, aggravated AngII-induced AAA formation in vivo. RNA-sequencing analysis of the suprarenal aortas of 4-day-AngII-infused ApoE-/- mice and bioinformatics analysis with ChIP-Atlas database identified the potential involvement of the osteogenic transcriptional factor Runx2 (runt-related transcription factor 2) in AAA. Consistently, vascular smooth muscle cell-specific Runx2 deficiency markedly repressed AngII-induced AAA formation in the ApoE-/- mice compared with the control littermates. CONCLUSIONS Our studies have revealed microcalcification as a novel pathological characteristic and potential mediator of AAA, and targeting microcalcification may represent a promising strategy for AAA prevention and treatment.
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, China (Z.L., Z.C., L.Y., Y.F., W.K.)
| | - Zuoquan Zhao
- Department of Nuclear Medicine (Z.Z., W.F.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, China (Z.L., Z.C., L.Y., Y.F., W.K.)
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham (Y.S., Y.C.)
| | - Li Li
- Department of Pathology, State Key Laboratory of Cardiovascular Disease (L.L.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Fang Yao
- State Key Laboratory of Cardiovascular Disease (F.Y., L.W.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Liu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, China (Z.L., Z.C., L.Y., Y.F., W.K.)
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, China (Y.Z.)
| | - Haibo Zhu
- Fuwai Hospital, National Center for Cardiovascular Diseases, and State Key Laboratory for Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica (H.Z.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, China (Z.L., Z.C., L.Y., Y.F., W.K.)
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease (F.Y., L.W.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Wei Fang
- Department of Nuclear Medicine (Z.Z., W.F.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham (Y.S., Y.C.)
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, China (Z.L., Z.C., L.Y., Y.F., W.K.)
| |
Collapse
|
164
|
Goody PR, Hosen MR, Christmann D, Niepmann ST, Zietzer A, Adam M, Bönner F, Zimmer S, Nickenig G, Jansen F. Aortic Valve Stenosis: From Basic Mechanisms to Novel Therapeutic Targets. Arterioscler Thromb Vasc Biol 2020; 40:885-900. [PMID: 32160774 DOI: 10.1161/atvbaha.119.313067] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aortic valve stenosis is the most prevalent heart valve disease worldwide. Although interventional treatment options have rapidly improved in recent years, symptomatic aortic valve stenosis is still associated with high morbidity and mortality. Calcific aortic valve stenosis is characterized by a progressive fibro-calcific remodeling and thickening of the aortic valve cusps, which subsequently leads to valve obstruction. The underlying pathophysiology is complex and involves endothelial dysfunction, immune cell infiltration, myofibroblastic and osteoblastic differentiation, and, subsequently, calcification. To date, no pharmacotherapy has been established to prevent aortic valve calcification. However, novel promising therapeutic targets have been recently identified. This review summarizes the current knowledge of pathomechanisms involved in aortic valve calcification and points out novel treatment strategies.
Collapse
Affiliation(s)
- Philip Roger Goody
- From the Heart Center Bonn, Department of Medicine II, University Hospital Bonn, Germany (P.R.G., M.R.H., D.C., S.T.N., S.Z., G.N., F.J.)
| | - Mohammed Rabiul Hosen
- From the Heart Center Bonn, Department of Medicine II, University Hospital Bonn, Germany (P.R.G., M.R.H., D.C., S.T.N., S.Z., G.N., F.J.)
| | - Dominik Christmann
- From the Heart Center Bonn, Department of Medicine II, University Hospital Bonn, Germany (P.R.G., M.R.H., D.C., S.T.N., S.Z., G.N., F.J.)
| | - Sven Thomas Niepmann
- From the Heart Center Bonn, Department of Medicine II, University Hospital Bonn, Germany (P.R.G., M.R.H., D.C., S.T.N., S.Z., G.N., F.J.)
| | | | - Matti Adam
- Clinic for Internal Medicine II, University Hospital Cologne, Germany (M.A.)
| | - Florian Bönner
- Clinic for Cardiology, Pulmonology, and Angiology, University Hospital Düsseldorf, Germany (F.B.)
| | - Sebastian Zimmer
- From the Heart Center Bonn, Department of Medicine II, University Hospital Bonn, Germany (P.R.G., M.R.H., D.C., S.T.N., S.Z., G.N., F.J.)
| | - Georg Nickenig
- From the Heart Center Bonn, Department of Medicine II, University Hospital Bonn, Germany (P.R.G., M.R.H., D.C., S.T.N., S.Z., G.N., F.J.)
| | - Felix Jansen
- From the Heart Center Bonn, Department of Medicine II, University Hospital Bonn, Germany (P.R.G., M.R.H., D.C., S.T.N., S.Z., G.N., F.J.)
| |
Collapse
|
165
|
Joshi NV, Elkhawad M, Forsythe RO, McBride OMB, Rajani NK, Tarkin JM, Chowdhury MM, Donoghue E, Robson JMJ, Boyle JR, Fryer TD, Huang Y, Teng Z, Dweck MR, Tawakol AA, Gillard JH, Coughlin PA, Wilkinson IB, Newby DE, Rudd JHF. Greater aortic inflammation and calcification in abdominal aortic aneurysmal disease than atherosclerosis: a prospective matched cohort study. Open Heart 2020; 7:e001141. [PMID: 32201583 PMCID: PMC7066636 DOI: 10.1136/openhrt-2019-001141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 01/06/2020] [Accepted: 01/21/2020] [Indexed: 01/24/2023] Open
Abstract
Objective Using combined positron emission tomography and CT (PET-CT), we measured aortic inflammation and calcification in patients with abdominal aortic aneurysms (AAA), and compared them with matched controls with atherosclerosis. Methods We prospectively recruited 63 patients (mean age 76.1±6.8 years) with asymptomatic aneurysm disease (mean size 4.33±0.73 cm) and 19 age-and-sex-matched patients with confirmed atherosclerosis but no aneurysm. Inflammation and calcification were assessed using combined 18F-FDG PET-CT and quantified using tissue-to-background ratios (TBRs) and Agatston scores. Results In patients with AAA, 18F-FDG uptake was higher within the aneurysm than in other regions of the aorta (mean TBRmax2.23±0.46 vs 2.12±0.46, p=0.02). Compared with atherosclerotic control subjects, both aneurysmal and non-aneurysmal aortae showed higher 18F-FDG accumulation (total aorta mean TBRmax2.16±0.51 vs 1.70±0.22, p=0.001; AAA mean TBRmax2.23±0.45 vs 1.68±0.21, p<0.0001). Aneurysms containing intraluminal thrombus demonstrated lower 18F-FDG uptake within their walls than those without (mean TBRmax2.14±0.43 vs 2.43±0.45, p=0.018), with thrombus itself showing low tracer uptake (mean TBRmax thrombus 1.30±0.48 vs aneurysm wall 2.23±0.46, p<0.0001). Calcification in the aneurysmal segment was higher than both non-aneurysmal segments in patients with aneurysm (Agatston 4918 (2901-8008) vs 1017 (139-2226), p<0.0001) and equivalent regions in control patients (442 (304-920) vs 166 (80-374) Agatston units per cm, p=0.0042). Conclusions The entire aorta is more inflamed in patients with aneurysm than in those with atherosclerosis, perhaps suggesting a generalised inflammatory aortopathy in patients with aneurysm. Calcification was prominent within the aneurysmal sac, with the remainder of the aorta being relatively spared. The presence of intraluminal thrombus, itself metabolically relatively inert, was associated with lower levels of inflammation in the adjacent aneurysmal wall.
Collapse
Affiliation(s)
- Nikhil V Joshi
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Maysoon Elkhawad
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Rachael O Forsythe
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Olivia M B McBride
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Nikil K Rajani
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Jason M Tarkin
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Mohammed M Chowdhury
- Department of Vascular Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Emma Donoghue
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Jonathan R Boyle
- Department of Vascular Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Yuan Huang
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Zhongzhao Teng
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Jonathan H Gillard
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Patrick A Coughlin
- Department of Vascular Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Ian B Wilkinson
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
166
|
Rogers MA, Chen J, Nallamshetty S, Pham T, Goto S, Muehlschlegel JD, Libby P, Aikawa M, Aikawa E, Plutzky J. Retinoids Repress Human Cardiovascular Cell Calcification With Evidence for Distinct Selective Retinoid Modulator Effects. Arterioscler Thromb Vasc Biol 2020; 40:656-669. [PMID: 31852220 PMCID: PMC7047603 DOI: 10.1161/atvbaha.119.313366] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Retinoic acid (RA) is a ligand for nuclear receptors that modulate gene transcription and cell differentiation. Whether RA controls ectopic calcification in humans is unknown. We tested the hypothesis that RA regulates osteogenic differentiation of human arterial smooth muscle cells and aortic valvular interstitial cells that participate in atherosclerosis and heart valve disease, respectively. Approach and Results: Human cardiovascular tissue contains immunoreactive RAR (RA receptor)-a retinoid-activated nuclear receptor directing multiple transcriptional programs. RA stimulation suppressed primary human cardiovascular cell calcification while treatment with the RAR inhibitor AGN 193109 or RARα siRNA increased calcification. RA attenuated calcification in a coordinated manner, increasing levels of the calcification inhibitor MGP (matrix Gla protein) while decreasing calcification-promoting TNAP (tissue nonspecific alkaline phosphatase) activity. Given that nuclear receptor action varies as a function of distinct ligand structures, we compared calcification responses to cyclic retinoids and the acyclic retinoid peretinoin. Peretinoin suppressed human cardiovascular cell calcification without inducing either secretion of APOC3 (apolipoprotein-CIII), which promotes atherogenesis, or reducing CYP7A1 (cytochrome P450 family 7 subfamily A member 1) expression, which occurred with cyclic retinoids all-trans RA, 9-cis RA, and 13-cis RA. Additionally, peretinoin did not suppress human femur osteoblast mineralization, whereas all-trans RA inhibited osteoblast mineralization. CONCLUSIONS These results establish retinoid regulation of human cardiovascular calcification, provide new insight into mechanisms involved in these responses, and suggest selective retinoid modulators, like acyclic retinoids may allow for treating cardiovascular calcification without the adverse effects associated with cyclic retinoids.
Collapse
MESH Headings
- Alkaline Phosphatase
- Aortic Valve/drug effects
- Aortic Valve/metabolism
- Aortic Valve/pathology
- Apolipoprotein C-III/genetics
- Apolipoprotein C-III/metabolism
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Carotid Arteries/drug effects
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Cholesterol 7-alpha-Hydroxylase/genetics
- Cholesterol 7-alpha-Hydroxylase/metabolism
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Heart Valve Diseases/genetics
- Heart Valve Diseases/metabolism
- Heart Valve Diseases/pathology
- Heart Valve Diseases/prevention & control
- Humans
- Isotretinoin/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Osteogenesis/drug effects
- Receptors, Retinoic Acid/agonists
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoids/pharmacology
- Retinoids/toxicity
- Signal Transduction
- Tretinoin/pharmacology
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/prevention & control
- Matrix Gla Protein
Collapse
Affiliation(s)
- Maximillian A. Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jiaohua Chen
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Shriram Nallamshetty
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Shinji Goto
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jochen D. Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Peter Libby
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jorge Plutzky
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| |
Collapse
|
167
|
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
|
168
|
Wang J, Chen X, Yang X, Guo B, Li D, Zhu X, Zhang X. Positive role of calcium phosphate ceramics regulated inflammation in the osteogenic differentiation of mesenchymal stem cells. J Biomed Mater Res A 2020; 108:1305-1320. [PMID: 32064734 DOI: 10.1002/jbm.a.36903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 02/05/2023]
Abstract
Recently, researches have confirmed the crucial role of inflammatory response in Ca-P ceramic-induced osteogenesis, however, the underlying mechanism has not yet been fully understood. In this study, BCP and β-TCP ceramics were used as material models to investigate the effect of physicochemical properties on inflammatory response in vitro. The results showed that BCP and β-TCP could support macrophages attachment, proliferation, and spreading favorably, as well as promote gene expressions of inflammatory related cytokines (IL-1, IL-6, MCP-1, and TNF-α) and growth factors (TGF-β, FGF, PDGF, VEGF, IGF, and EGF). BCP showed a facilitating function on the gene expressions earlier than β-TCP. Further coculture experiments performed in vitro demonstrated that the CMs containing various increased cytokines for macrophages pre-culture could significantly promote MSCs osteogenic differentiation, which was confirmed by the gene expressions of osteogenic specific markers and the intracellular OCN product accumulation under the stimulation of BCP and β-TCP ceramics. Further evidence was found from the formation of mineralized nodules in BCM and TCM. In addition, this study showed a concise relationship between Ca-P ceramic induced inflammation and its osteoinductivity that the increased cytokines and growth factors from macrophages could promote MSCs osteogenic differentiation.
Collapse
Affiliation(s)
- Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xuening Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Bo Guo
- Department of Ophthalmology, West China Hospital of Sichuan University, Chengdu, China
| | - Danyang Li
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
169
|
Shioi A, Morioka T, Shoji T, Emoto M. The Inhibitory Roles of Vitamin K in Progression of Vascular Calcification. Nutrients 2020; 12:nu12020583. [PMID: 32102248 PMCID: PMC7071387 DOI: 10.3390/nu12020583] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Vitamin K is a fat-soluble vitamin that is indispensable for the activation of vitamin K-dependent proteins (VKDPs) and may be implicated in cardiovascular disease (CVD). Vascular calcification is intimately associated with CV events and mortality and is a chronic inflammatory process in which activated macrophages promote osteoblastic differentiation of vascular smooth muscle cells (VSMCs) through the production of proinflammatory cytokines such as IL-1β, IL-6, TNF-α, and oncostatin M (OSM) in both intimal and medial layers of arterial walls. This process may be mainly mediated through NF-κB signaling pathway. Vitamin K has been demonstrated to exert anti-inflammatory effects through antagonizing NF-κB signaling in both in vitro and in vivo studies, suggesting that vitamin K may prevent vascular calcification via anti-inflammatory mechanisms. Matrix Gla protein (MGP) is a major inhibitor of soft tissue calcification and contributes to preventing both intimal and medial vascular calcification. Vitamin K may also inhibit progression of vascular calcification by enhancing the activity of MGP through facilitating its γ-carboxylation. In support of this hypothesis, the procalcific effects of warfarin, an antagonist of vitamin K, on arterial calcification have been demonstrated in several clinical studies. Among the inactive MGP forms, dephospho-uncarboxylated MGP (dp-ucMGP) may be regarded as the most useful biomarker of not only vitamin K deficiency, but also vascular calcification and CVD. There have been several studies showing the association of circulating levels of dp-ucMGP with vitamin K intake, vascular calcification, mortality, and CVD. However, additional larger prospective studies including randomized controlled trials are necessary to confirm the beneficial effects of vitamin K supplementation on CV health.
Collapse
Affiliation(s)
- Atsushi Shioi
- Department of Vascular Medicine and Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan;
- Correspondence: ; Tel.: +81666453931
| | - Tomoaki Morioka
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka 545-85858, Japan; (T.M.); (M.E.)
| | - Tetsuo Shoji
- Department of Vascular Medicine and Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan;
| | - Masanori Emoto
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka 545-85858, Japan; (T.M.); (M.E.)
| |
Collapse
|
170
|
Elagin V, Kuznetsova D, Grebenik E, Zolotov DA, Istranov L, Zharikova T, Istranova E, Polozova A, Reunov D, Kurkov A, Shekhter A, Gafarova ER, Asadchikov V, Borisov SM, Dmitriev RI, Zagaynova E, Timashev P. Multiparametric Optical Bioimaging Reveals the Fate of Epoxy Crosslinked Biomeshes in the Mouse Subcutaneous Implantation Model. Front Bioeng Biotechnol 2020; 8:107. [PMID: 32140465 PMCID: PMC7042178 DOI: 10.3389/fbioe.2020.00107] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 02/03/2020] [Indexed: 12/13/2022] Open
Abstract
Biomeshes based on decellularized bovine pericardium (DBP) are widely used in reconstructive surgery due to their wide availability and the attractive biomechanical properties. However, their efficacy in clinical applications is often affected by the uncontrolled immunogenicity and proteolytic degradation. To address this issue, we present here in vivo multiparametric imaging analysis of epoxy crosslinked DBPs to reveal their fate after implantation. We first analyzed the structure of the crosslinked DBP using scanning electron microscopy and evaluated proteolytic stability and cytotoxicity. Next, using combination of fluorescence and hypoxia imaging, X-ray computed microtomography and histology techniques we studied the fate of DBPs after subcutaneous implantation in animals. Our approach revealed high resistance to biodegradation, gradual remodeling of a surrounding tissue forming the connective tissue capsule and calcification of crosslinked DBPs. These changes were concomitant to the development of hypoxia in the samples within 3 weeks after implantation and subsequent induction of angiogenesis and vascularization. Collectively, presented approach provides new insights on the transplantation of the epoxy crosslinked biomeshes, the risks associated with its applications in soft-tissue reconstruction and can be transferred to studies of other types of implants.
Collapse
Affiliation(s)
- Vadim Elagin
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - Daria Kuznetsova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - Ekaterina Grebenik
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Denis A Zolotov
- Shubnikov Institute of Crystallography, Federal Scientific Research Centre "Crystallography and Photonics" Russian Academy of Sciences, Moscow, Russia
| | - Leonid Istranov
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Tatiana Zharikova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Elena Istranova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anastasia Polozova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - Dmitry Reunov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - Alexandr Kurkov
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anatoly Shekhter
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Elvira R Gafarova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Victor Asadchikov
- Shubnikov Institute of Crystallography, Federal Scientific Research Centre "Crystallography and Photonics" Russian Academy of Sciences, Moscow, Russia
| | - Sergey M Borisov
- Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology, Graz, Austria
| | - Ruslan I Dmitriev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Elena Zagaynova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Photonic Technologies, Federal Scientific Research Centre "Crystallography and Photonics" Russian Academy of Sciences, Moscow, Russia.,Department of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, Moscow, Russia
| |
Collapse
|
171
|
Guaraldi G, Milic J, Prandini N, Ligabue G, Esposito F, Ciusa G, Malagoli A, Scaglioni R, Besutti G, Beghetto B, Nardini G, Roncaglia E, Mussini C, Raggi P. 18Fluoride-based molecular imaging of coronary atherosclerosis in HIV infected patients. Atherosclerosis 2020; 297:127-135. [PMID: 32113050 DOI: 10.1016/j.atherosclerosis.2020.02.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/18/2020] [Accepted: 02/14/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Molecular imaging with 18Fluorodeoxyglucose (FDG) and 18F-sodium-fluoride (NaF) captures arterial inflammation and micro-calcification and can reveal potentially unstable atherosclerotic plaques. METHODS We performed FDG and NaF PET/CT imaging in two clinically similar cohorts of patients living with HIV (PLWH) with no symptomatic cardiovascular disease. The prevalence and intensity of coronary artery uptake of each tracer, measured as target-to-background ratio (TBR), were assessed in patients at low and high cardiovascular risk. RESULTS Ninety-three PLWH were submitted to PET/CT imaging with FDG (N = 43) and NaF (N = 50); 42% were at low and 58% at high cardiovascular risk. The intensity of uptake and multivessel coronary artery uptake were significantly higher with NaF than FDG both in low and high-risk patients. When each 18F-tracer was tested in low and high-risk patients, an equal proportion of subjects showed no vessel, single and multivessel NaF uptake; the same was true for no and single vessel uptake of FDG (no multivessel FDG uptake was noted). Waist circumference, CRP, D-dimer, HIV duration and treatment with nucleoside reverse transcriptase inhibitors were associated with high NaF uptake in univariable analyses; D-dimer remained significant in multivariable analyses (OR = 1.05; p=0.02). There were no significant associations with FDG uptake. CONCLUSIONS The prevalence of coronary artery uptake was higher with NaF compared to FDG both in high and low risk patients, hence microcalcification imaging may be a more sensitive tool to detect coronary atherosclerosis than inflammation imaging. However, the uptake of each 18Fluoride tracer was similar between low and high-risk subjects, and this underscores the discordance between clinical and imaging based risk assessment. Future investigation should address the prognostic significance of NaF coronary artery uptake.
Collapse
Affiliation(s)
- Giovanni Guaraldi
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Jovana Milic
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Napoleone Prandini
- Department of Nuclear Medicine, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Guido Ligabue
- Department of Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Francesco Esposito
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Giacomo Ciusa
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Andrea Malagoli
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Riccardo Scaglioni
- Department of Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Giulia Besutti
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41124, Modena, Italy; Department of Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Barbara Beghetto
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Giulia Nardini
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Enrica Roncaglia
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Cristina Mussini
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Paolo Raggi
- Division of Cardiology and Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, 11220, 83rd Avenue, Suite 5A9-014, Edmonton, AB T6G 2B7, Canada.
| |
Collapse
|
172
|
Shi X, Gao J, Lv Q, Cai H, Wang F, Ye R, Liu X. Calcification in Atherosclerotic Plaque Vulnerability: Friend or Foe? Front Physiol 2020; 11:56. [PMID: 32116766 PMCID: PMC7013039 DOI: 10.3389/fphys.2020.00056] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
Calcification is a clinical marker of atherosclerosis. This review focuses on recent findings on the association between calcification and plaque vulnerability. Calcified plaques have traditionally been regarded as stable atheromas, those causing stenosis may be more stable than non-calcified plaques. With the advances in intravascular imaging technology, the detection of the calcification and its surrounding plaque components have evolved. Microcalcifications and spotty calcifications represent an active stage of vascular calcification correlated with inflammation, whereas the degree of plaque calcification is strongly inversely related to macrophage infiltration. Asymptomatic patients have a higher content of plaque calcification than that in symptomatic patients. The effect of calcification might be biphasic. Plaque rupture has been shown to correlate positively with the number of spotty calcifications, and inversely with the number of large calcifications. There may be certain stages of calcium deposition that may be more atherogenic. Moreover, superficial calcifications are independently associated with plaque rupture and intraplaque hemorrhage, which may be due to the concentrated and asymmetrical distribution of biological stress in plaques. Conclusively, calcification of differential amounts, sizes, shapes, and positions may play differential roles in plaque homeostasis. The surrounding environments around the calcification within plaques also have impacts on plaque homeostasis. The interactive effects of these important factors of calcifications and plaques still await further study.
Collapse
Affiliation(s)
- Xuan Shi
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Jie Gao
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Qiushi Lv
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Haodi Cai
- Department of Neurology, Jinling Hospital, Southeast University, Nanjing, China
| | - Fang Wang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ruidong Ye
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
173
|
Healy A, Berus JM, Christensen JL, Lee C, Mantsounga C, Dong W, Watts JP, Assali M, Ceneri N, Nilson R, Neverson J, Wu WC, Choudhary G, Morrison AR. Statins Disrupt Macrophage Rac1 Regulation Leading to Increased Atherosclerotic Plaque Calcification. Arterioscler Thromb Vasc Biol 2020; 40:714-732. [PMID: 31996022 DOI: 10.1161/atvbaha.119.313832] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Calcification of atherosclerotic plaque is traditionally associated with increased cardiovascular event risk; however, recent studies have found increased calcium density to be associated with more stable disease. 3-hydroxy-3-methylglutaryl coenzymeA reductase inhibitors or statins reduce cardiovascular events. Invasive clinical studies have found that statins alter both the lipid and calcium composition of plaque but the molecular mechanisms of statin-mediated effects on plaque calcium composition remain unclear. We recently defined a macrophage Rac (Ras-related C3 botulinum toxin substrate)-IL-1β (interleukin-1 beta) signaling axis to be a key mechanism in promoting atherosclerotic calcification and sought to define the impact of statin therapy on this pathway. Approach and Results: Here, we demonstrate that statin therapy is independently associated with elevated coronary calcification in a high-risk patient population and that statins disrupt the complex between Rac1 and its inhibitor RhoGDI (Rho GDP-dissociation inhibitor), leading to increased active (GTP bound) Rac1 in primary monocytes/macrophages. Rac1 activation is prevented by rescue with the isoprenyl precursor geranylgeranyl diphosphate. Statin-treated macrophages exhibit increased activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), increased IL-1β mRNA, and increased Rac1-dependent IL-1β protein secretion in response to inflammasome stimulation. Using an animal model of calcific atherosclerosis, inclusion of statin in the atherogenic diet led to a myeloid Rac1-dependent increase in atherosclerotic calcification, which was associated with increased serum IL-1β expression, increased plaque Rac1 activation, and increased plaque expression of the osteogenic markers, alkaline phosphatase and RUNX2 (Runt-related transcription factor 2). CONCLUSIONS Statins are capable of increasing atherosclerotic calcification through disinhibition of a macrophage Rac1-IL-1β signaling axis.
Collapse
Affiliation(s)
- Abigail Healy
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Joshua M Berus
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Jared L Christensen
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Cadence Lee
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Chris Mantsounga
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Willie Dong
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Jerome P Watts
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Maen Assali
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Nicolle Ceneri
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Rachael Nilson
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Jade Neverson
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Wen-Chih Wu
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Gaurav Choudhary
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Alan R Morrison
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| |
Collapse
|
174
|
Karpouzas GA, Ormseth SR, Hernandez E, Budoff MJ. Impact of Cumulative Inflammation, Cardiac Risk Factors, and Medication Exposure on Coronary Atherosclerosis Progression in Rheumatoid Arthritis. Arthritis Rheumatol 2020; 72:400-408. [PMID: 31532064 DOI: 10.1002/art.41122] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 09/12/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To explore incidence and progression of coronary atherosclerosis and identify determinants in patients with rheumatoid arthritis (RA). We specifically evaluated the impact of inflammation, cardiac risk factors, duration of medication exposure, and their interactions on coronary plaque progression. METHODS One hundred one participants with baseline coronary computed tomography angiography findings underwent follow-up assessment a mean ± SD of 83 ± 3.6 months after baseline. Plaque burden was reported as the segment involvement score (describing the number of coronary segments with plaque) and the segment stenosis score (characterizing the cumulative plaque stenosis over all evaluable segments). Plaque composition was classified as noncalcified, mixed, or calcified. Coronary artery calcium (CAC) was quantified using the Agatston method. RESULTS Total plaque increased in 48% of patients, and progression was predicted by older age, higher cumulative inflammation, and total prednisone dose (P < 0.05). CAC progressors were older, more obese, hypertensive, and had higher cumulative inflammation compared to nonprogressors (P < 0.05). Longer exposure to biologics was associated with lower likelihood of noncalcified plaque progression, lesion remodeling, and constrained CAC change in patients without baseline calcification, independent of inflammation, prednisone dose, or statin exposure (all P < 0.05). Longer statin treatment further restricted noncalcified plaque progression and attenuated the effect of inflammation on increased plaque and CAC (P < 0.05). Stringent systolic blood pressure (BP) control further weakened the effect of inflammation on total plaque progression. CONCLUSION Inflammation was a consistent and independent predictor of coronary atherosclerosis progression in RA. It should therefore be specifically targeted toward mitigating cardiovascular risk. Biologic disease-modifying antirheumatic drugs, statins, and BP control may further constrain plaque progression directly or indirectly.
Collapse
|
175
|
Rogers MA, Aikawa E. Cardiovascular calcification: artificial intelligence and big data accelerate mechanistic discovery. Nat Rev Cardiol 2020; 16:261-274. [PMID: 30531869 DOI: 10.1038/s41569-018-0123-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular calcification is a health disorder with increasing prevalence and high morbidity and mortality. The only available therapeutic options for calcific vascular and valvular heart disease are invasive transcatheter procedures or surgeries that do not fully address the wide spectrum of these conditions; therefore, an urgent need exists for medical options. Cardiovascular calcification is an active process, which provides a potential opportunity for effective therapeutic targeting. Numerous biological processes are involved in calcific disease, including matrix remodelling, transcriptional regulation, mitochondrial dysfunction, oxidative stress, calcium and phosphate signalling, endoplasmic reticulum stress, lipid and mineral metabolism, autophagy, inflammation, apoptosis, loss of mineralization inhibition, impaired mineral resorption, cellular senescence and extracellular vesicles that act as precursors of microcalcification. Advances in molecular imaging and big data technology, including in multiomics and network medicine, and the integration of these approaches are helping to provide a more comprehensive map of human disease. In this Review, we discuss ectopic calcification processes in the cardiovascular system, with an emphasis on emerging mechanistic knowledge obtained through patient data and advances in imaging methods, experimental models and multiomics-generated big data. We also highlight the potential and challenges of artificial intelligence, machine learning and deep learning to integrate imaging and mechanistic data for drug discovery.
Collapse
Affiliation(s)
- Maximillian A Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
176
|
Chen TC, Yen CK, Lu YC, Shi CS, Hsieh RZ, Chang SF, Chen CN. The antagonism of 6-shogaol in high-glucose-activated NLRP3 inflammasome and consequent calcification of human artery smooth muscle cells. Cell Biosci 2020; 10:5. [PMID: 31938471 PMCID: PMC6953308 DOI: 10.1186/s13578-019-0372-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
Background Vascular calcification is the major reason for high mortality of cardiovascular complications for diabetes. Interleukin (IL)-1β has been implicated in this pathogenesis, but its precise role and clinical evidence have not been clearly identified. Hence, this study was aimed to investigate whether high concentration of glucose (HG), which mimics the hyperglycemia environment, could initiate vascular calcification through NLRP3/IL-1β inflammasome and the underlying mechanism. Recently, 6-shogaol, a major ginger derivate, has been elucidated its pharmaceutic role for various diseases. Therefore, the aims of this study also determined 6-shogaol effect in vascular calcification of HG initiation. Result Human artery smooth muscle cells (HASMCs) were used in this study. Glucose concentrations at 5 and 25 mM were defined as normal and HG status, respectively. The results showed that HG could increase the NLRP3, cleaved caspase 1, and pro/mature IL-1β levels to induce the expressions of bone-related matrix proteins and subsequent HASMC calcification. This process was regulated by Akt activation and reactive oxygen species (ROS) production. Moreover, 6-shogaol could inhibit the Akt/ROS signaling and NLRP3/caspase 1/IL-1β inflammasome and hence attenuated HASMC calcification. Conclusions This study elucidates the detailed mechanism of HG-initiated HASMC calcification through NLRP3/caspase 1/IL-1β inflammasome and indicates a potential therapeutic role of 6-shogaol in vascular calcification complication of diabetes.
Collapse
Affiliation(s)
- Te-Chuan Chen
- 1Division of Nephrology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Kung Yen
- 2Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Ying-Chen Lu
- 2Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Chung-Sheng Shi
- 3Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,4Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Rong-Ze Hsieh
- 3Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,5Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Shun-Fu Chang
- 5Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Cheng-Nan Chen
- 6Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 600 Taiwan
| |
Collapse
|
177
|
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
|
178
|
Relationship between coronary arterial 18F-sodium fluoride uptake and epicardial adipose tissue analyzed using computed tomography. Eur J Nucl Med Mol Imaging 2020; 47:1746-1756. [PMID: 31897585 DOI: 10.1007/s00259-019-04675-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/26/2019] [Indexed: 02/08/2023]
Abstract
PURPOSE 18F-Sodium fluoride (18F-NaF) positron emission tomography (PET) has the potential to detect high-risk coronary plaques. Epicardial adipose tissue (EAT) reportedly correlates with coronary atherosclerosis progression. We evaluated the relationship between coronary arterial 18F-NaF uptake and EAT findings using computed tomography (CT). METHODS We studied 40 patients with ≥ 1 coronary plaque detected on cardiac CT who underwent 18F-NaF PET/CT. EAT volume was measured using CT and indexed to body surface area in each patient. Each plaque was evaluated for CT-based luminal stenosis and high-risk features. The mean EAT density surrounding each plaque was calculated as perilesional EAT density (PLED) using non-contrast CT images. Focal 18F-NaF uptake in each plaque was quantified using the maximum tissue-to-background ratio (TBRmax). RESULTS EAT volume index was similar between patients with TBRmax ≥ 1.28 (previously reported optimal cutoff to predict coronary events) and those with lower TBRmax, but patients with TBRmax ≥ 1.28 showed higher maximum PLED per patient (- 86 ± 12 Hounsfield units (HU) versus - 98 ± 11 HU, P = 0.0044). In the lesion-based analysis (n = 92), PLED was positively correlated with TBRmax, and the optimal PLED cutoff to identify TBRmax ≥ 1.28 was - 97 HU. On multivariate analysis adjusted for lesion location, obstructive stenosis, and high-risk plaque on CT, PLED ≥ - 97 HU remained a significant predictor of TBRmax ≥ 1.28. CONCLUSIONS Increased PLED was associated with significant coronary arterial 18F-NaF uptake. Step-by-step analyses of EAT density on CT and coronary arterial 18F-NaF uptake on PET may offer novel strategies for risk prediction in coronary artery disease.
Collapse
|
179
|
|
180
|
Zarb Y, Weber-Stadlbauer U, Kirschenbaum D, Kindler DR, Richetto J, Keller D, Rademakers R, Dickson DW, Pasch A, Byzova T, Nahar K, Voigt FF, Helmchen F, Boss A, Aguzzi A, Klohs J, Keller A. Ossified blood vessels in primary familial brain calcification elicit a neurotoxic astrocyte response. Brain 2019; 142:885-902. [PMID: 30805583 PMCID: PMC6439320 DOI: 10.1093/brain/awz032] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/07/2018] [Accepted: 12/26/2018] [Indexed: 12/17/2022] Open
Abstract
Brain calcifications are commonly detected in aged individuals and accompany numerous brain diseases, but their functional importance is not understood. In cases of primary familial brain calcification, an autosomally inherited neuropsychiatric disorder, the presence of bilateral brain calcifications in the absence of secondary causes of brain calcification is a diagnostic criterion. To date, mutations in five genes including solute carrier 20 member 2 (SLC20A2), xenotropic and polytropic retrovirus receptor 1 (XPR1), myogenesis regulating glycosidase (MYORG), platelet-derived growth factor B (PDGFB) and platelet-derived growth factor receptor β (PDGFRB), are considered causal. Previously, we have reported that mutations in PDGFB in humans are associated with primary familial brain calcification, and mice hypomorphic for PDGFB (Pdgfbret/ret) present with brain vessel calcifications in the deep regions of the brain that increase with age, mimicking the pathology observed in human mutation carriers. In this study, we characterize the cellular environment surrounding calcifications in Pdgfbret/ret animals and show that cells around vessel-associated calcifications express markers for osteoblasts, osteoclasts and osteocytes, and that bone matrix proteins are present in vessel-associated calcifications. Additionally, we also demonstrate the osteogenic environment around brain calcifications in genetically confirmed primary familial brain calcification cases. We show that calcifications cause oxidative stress in astrocytes and evoke expression of neurotoxic astrocyte markers. Similar to previously reported human primary familial brain calcification cases, we describe high interindividual variation in calcification load in Pdgfbret/ret animals, as assessed by ex vivo and in vivo quantification of calcifications. We also report that serum of Pdgfbret/ret animals does not differ in calcification propensity from control animals and that vessel calcification occurs only in the brains of Pdgfbret/ret animals. Notably, ossification of vessels and astrocytic neurotoxic response is associated with specific behavioural and cognitive alterations, some of which are associated with primary familial brain calcification in a subset of patients.
Collapse
Affiliation(s)
- Yvette Zarb
- Department of Neurosurgery, Clinical Neuroscience Center, Zurich University Hospital, Zurich University, Zurich, Switzerland.,Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich University, Zurich, Switzerland
| | - Daniel Kirschenbaum
- Department of Neurosurgery, Clinical Neuroscience Center, Zurich University Hospital, Zurich University, Zurich, Switzerland
| | - Diana Rita Kindler
- Institute of Neuropathology, Zurich University Hospital, Zurich University, Zurich, Switzerland
| | - Juliet Richetto
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich University, Zurich, Switzerland
| | - Daniel Keller
- Department of Biomedical Engineering, ETH and University of Zurich, Zurich, Switzerland
| | - Rosa Rademakers
- Institute of Diagnostic and Interventional Radiology, Zurich University Hospital, Zurich University, Zurich, Switzerland
| | - Dennis W Dickson
- Institute of Diagnostic and Interventional Radiology, Zurich University Hospital, Zurich University, Zurich, Switzerland
| | - Andreas Pasch
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Khayrun Nahar
- Department of Neurosurgery, Clinical Neuroscience Center, Zurich University Hospital, Zurich University, Zurich, Switzerland
| | - Fabian F Voigt
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Brain Research Institute, Zurich University, Zurich, Switzerland
| | - Fritjof Helmchen
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Brain Research Institute, Zurich University, Zurich, Switzerland
| | - Andreas Boss
- Department of Biomedical Engineering, ETH and University of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Department of Neurosurgery, Clinical Neuroscience Center, Zurich University Hospital, Zurich University, Zurich, Switzerland
| | - Jan Klohs
- Institute of Neuropathology, Zurich University Hospital, Zurich University, Zurich, Switzerland
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, Zurich University Hospital, Zurich University, Zurich, Switzerland.,Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
181
|
Coronary Atherosclerosis in Masters Athletes: Mechanisms and Implications for Cardiovascular Disease Risk. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2019; 21:87. [DOI: 10.1007/s11936-019-0798-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
182
|
Malhotra R, Wunderer F, Barnes HJ, Bagchi A, Buswell MD, O'Rourke CD, Slocum CL, Ledsky CD, Peneyra KM, Sigurslid H, Corman B, Johansson KB, Rhee DK, Bloch KD, Bloch DB. Hepcidin Deficiency Protects Against Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 39:178-187. [PMID: 30587002 DOI: 10.1161/atvbaha.118.312215] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective- Inflammatory stimuli enhance the progression of atherosclerotic disease. Inflammation also increases the expression of hepcidin, a hormonal regulator of iron homeostasis, which decreases intestinal iron absorption, reduces serum iron levels and traps iron within macrophages. The role of macrophage iron in the development of atherosclerosis remains incompletely understood. The objective of this study was to investigate the effects of hepcidin deficiency and decreased macrophage iron on the development of atherosclerosis. Approach and Results- Hepcidin- and LDL (low-density lipoprotein) receptor-deficient ( Hamp-/-/ Ldlr-/-) mice and Hamp+/+/ Ldlr-/- control mice were fed a high-fat diet for 21 weeks. Compared with control mice, Hamp-/-/ Ldlr-/- mice had decreased aortic macrophage activity and atherosclerosis. Because hepcidin deficiency is associated with both increased serum iron and decreased macrophage iron, the possibility that increased serum iron was responsible for decreased atherosclerosis in Hamp-/-/ Ldlr-/- mice was considered. Hamp+/+/ Ldlr-/- mice were treated with iron dextran so as to produce a 2-fold increase in serum iron. Increased serum iron did not decrease atherosclerosis in Hamp+/+/ Ldlr-/- mice. Aortic macrophages from Hamp-/-/ Ldlr-/- mice had less labile free iron and exhibited a reduced proinflammatory (M1) phenotype compared with macrophages from Hamp+/+/ Ldlr-/- mice. THP1 human macrophages treated with an iron chelator were used to model hepcidin deficiency in vitro. Treatment with an iron chelator reduced LPS (lipopolysaccharide)-induced M1 phenotypic expression and decreased uptake of oxidized LDL. Conclusions- In summary, in a hyperlipidemic mouse model, hepcidin deficiency was associated with decreased macrophage iron, a reduced aortic macrophage inflammatory phenotype and protection from atherosclerosis. The results indicate that decreasing hepcidin activity, with the resulting decrease in macrophage iron, may prove to be a novel strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Rajeev Malhotra
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Florian Wunderer
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston.,Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt am Main, Germany (F.W.)
| | - Hanna J Barnes
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Aranya Bagchi
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Mary D Buswell
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Caitlin D O'Rourke
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Charles L Slocum
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Clara D Ledsky
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kathryn M Peneyra
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Haakon Sigurslid
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Benjamin Corman
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kimberly B Johansson
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - David K Rhee
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kenneth D Bloch
- From the Cardiovascular Research Center and Cardiology Division of the Department of Medicine (R.M., H.J.B., M.D.B., C.L.S., H.S., D.K.R., K.D.B.), Massachusetts General Hospital and Harvard Medical School, Boston.,the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Donald B Bloch
- the Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine (F.W., A.B., C.D.O., C.D.L., K.M.P., B.C., K.B.J., K.D.B., D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston.,Division of Rheumatology, Allergy and Immunology of the Department of Medicine (D.B.B.), Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
183
|
Scimeca M, Anemona L, Granaglia A, Bonfiglio R, Urbano N, Toschi N, Santeusanio G, Schiaroli S, Mauriello S, Tancredi V, Schillaci O, Bonanno E, Mauriello A. Plaque calcification is driven by different mechanisms of mineralization associated with specific cardiovascular risk factors. Nutr Metab Cardiovasc Dis 2019; 29:1330-1336. [PMID: 31653516 DOI: 10.1016/j.numecd.2019.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 08/01/2019] [Accepted: 08/14/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS The aim of this study was to investigate possible associations among markers of mineralization, plaque instability and the main risk factors of atherosclerosis. METHODS AND RESULTS A Tissue MicroArray containing 52 samples of calcified carotid plaques from 52 symptomatic and asymptomatic patients were built. TMA serial sections were used to study the expression of inflammatory and mineralization markers (BMP-2, BMP-4, VDR, RANKL, Osteopontin, Sclerostin, β-catenin and calmodulin) by immunohistochemistry. Our data clearly demonstrated the expression of mineralization markers in atheromatic plaques. Indeed, with the exception of RANKL, all investigated markers were expressed in at least 60% of cases. Specifically, multivariate analysis displayed significant associations between both the expression of BMP-2 and the presence of unstable plaques as well as between the expression of β-catenin and the presence of stable plaques. We also found a significant inverse association between both a) the presence of hypertension and VDR and b) smoking habits and calmodulin expression. Finally, we noted a higher density of RANKL positive cells in plaques from diabetic patients as compared to non-diabetic ones and a significant positive association between hypertriglyceridemia and BMP-4 expression. CONCLUSION Our results support the hypothesis that the process of atherosclerotic plaque calcification presents a number of similarities with the physiological processes that occur in bone, involving both osteoblasts- and osteoclasts-like arterial cells. Finally, the present study suggests that risk factors, such as hypertension, cigarette smoke and diabetes, can cause the destabilization of the atheromatic plaque acting on calcification process as well as inflammation.
Collapse
Affiliation(s)
- Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome, 00133, Italy; San Raffaele University, Via di Val Cannuta 247, 00166, Rome, Italy; Fondazione Umberto Veronesi (FUV), Piazza Velasca 5, 20122, Milano, Italy; Saint Camillus International University of Health Sciences, Via di Sant'Alessandro, 8, 00131 Rome, Italy.
| | - Lucia Anemona
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Annarita Granaglia
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Rita Bonfiglio
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | | | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome, 00133, Italy; Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA.
| | - Giuseppe Santeusanio
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Stefania Schiaroli
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Silvestro Mauriello
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome, 00133, Italy.
| | - Virginia Tancredi
- San Raffaele University, Via di Val Cannuta 247, 00166, Rome, Italy; Department of Systems Medicine, School of Sport and Exercise Sciences, University of Rome Tor Vergata, Rome, Italy; Centre of Space Biomedicine, University of Rome Tor Vergata, Rome, Italy.
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome, 00133, Italy; IRCCS Neuromed, Pozzilli, Italy.
| | - Elena Bonanno
- Saint Camillus International University of Health Sciences, Via di Sant'Alessandro, 8, 00131 Rome, Italy; IRCCS Neuromed Lab. "Diagnostica Medica"; and "Villa dei Platani", Avellino, Italy.
| | - Alessandro Mauriello
- Saint Camillus International University of Health Sciences, Via di Sant'Alessandro, 8, 00131 Rome, Italy; TorVergata Oncoscience Research (TOR), University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
184
|
Chin DD, Wang J, Mel de Fontenay M, Plotkin A, Magee GA, Chung EJ. Hydroxyapatite-binding micelles for the detection of vascular calcification in atherosclerosis. J Mater Chem B 2019; 7:6449-6457. [PMID: 31553027 PMCID: PMC6812598 DOI: 10.1039/c9tb01918a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a chronic disease characterized by the formation of calcified, arterial plaques. Microcalcifications (5 μm to 100 μm), mainly composed of hydroxyapatite (HA, Ca5(PO4)3(OH)), develop in the fibrous caps of atherosclerotic plaques and can trigger plaque rupture due to the loss of compliance and elasticity. Ultimately, plaque rupture can cause arterial occlusion and embolization and result in ischemic events such as strokes and myocardial infarctions. Unfortunately, current imaging technologies used to detect calcifications are limited by low signal-to-noise ratio or use invasive procedures that pose risk of arterial dissection. To mitigate these drawbacks, in our study, we developed a novel, fluorescently-labeled peptide amphiphile micelle (PAM) that uses a 12 amino acid HA-binding peptide (HABP) [SVSVGMKPSPRP] to target and detect atherosclerotic calcification (HA PAM). Our results show HA PAMs can successfully target HA microcrystals with a strong binding affinity (KD = 6.26 ± 1.2 μM) in vitro. In addition, HA PAMs detected HA mineralization (HA PAM vs. non-targeting micelle, p≤ 0.001; HA PAM vs. scrambled HABP PAM, p≤ 0.01) formed by calcifying mouse aortic vascular smooth muscle cells (MOVAS). Moreover, HA PAMs successfully detected calcifications in atherosclerotic mouse models as well as in patient-derived arteries. Our studies show that HA PAMs show promise as calcium-targeting nanoparticles for the detection of calcifications in atherosclerosis.
Collapse
Affiliation(s)
- Deborah D Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Margot Mel de Fontenay
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Anastasia Plotkin
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gregory A Magee
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA. and Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA and Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
185
|
Ren JL, Hou YL, Ni XQ, Zhu Q, Chen Y, Zhang LS, Liu X, Xue CD, Wu N, Yu YR, Tang CS, Ning ZP, Chai SB, Qi YF. Intermedin1-53 Ameliorates Homocysteine-Promoted Atherosclerotic Calcification by Inhibiting Endoplasmic Reticulum Stress. J Cardiovasc Pharmacol Ther 2019; 25:251-264. [DOI: 10.1177/1074248419885633] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Aim: Vascular calcification (VC) is thought to be an independent predictor of cardiovascular morbidity and mortality. Intermedin1-53 (IMD) is a cardiovascular protective peptide and can inhibit vascular medial calcification in rats. In this study, we investigated the effect of IMD on atherosclerotic calcification induced by a high-fat diet plus homocysteine (Hcy) and the potential mechanisms. Methods: ApoE−/− mice were fed a high-fat diet with Hcy in drinking water to induce atherosclerotic calcification. Results: As compared to the high-fat diet alone, Hcy treatment significantly increased atherosclerotic lesion areas and the number of calcified nodules in aortic roots and was reduced by IMD infusion or 4-phenylbutyric acid (PBA) treatment. In vitro, as compared to calcifying medium alone, Hcy treatment further increased alkaline phosphatase activity, calcium content, and calcium nodule number in human aorta vascular smooth muscle cells (HA-VSMCs), all blocked by IMD or PBA pretreatment. Mechanistically, IMD or PBA significantly alleviated endoplasmic reticulum stress (ERS) activation compared with Hcy treatment. In parallel, IMD or PBA attenuated the messenger RNA levels of osteogenic markers and inflammatory cytokines in aortas and their protein levels in lesions of aortic roots. In vitro, Hcy treatment significantly increased the protein levels of osteoblast-like cell markers in primary rat VSMCs and inflammation markers in mouse peritoneal macrophages, all decreased with IMD or PBA pretreatment. Intermedin1-53 pretreatment also markedly reduced the protein levels of ERS markers in rat VSMCs and mouse peritoneal macrophages. Conclusions: Intermedin1-53 protects against Hcy-promoted atherosclerotic calcification in ApoE−/− mice by inhibiting ERS.
Collapse
Affiliation(s)
- Jin-Ling Ren
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yue-Long Hou
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xian-Qiang Ni
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qing Zhu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yao Chen
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lin-Shuang Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xin Liu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chang-Ding Xue
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ning Wu
- Department of Gynaecology and Obstetrics, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yan-Rong Yu
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chao-Shu Tang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhong-Ping Ning
- Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - San-Bao Chai
- Department of Endocrinology, Peking University International Hospital, Beijing, China
| | - Yong-Fen Qi
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
- Department of Pathogen Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
186
|
Tucureanu MM, Filippi A, Alexandru N, Ana Constantinescu C, Ciortan L, Macarie R, Vadana M, Voicu G, Frunza S, Nistor D, Simionescu A, Simionescu DT, Georgescu A, Manduteanu I. Diabetes-induced early molecular and functional changes in aortic heart valves in a murine model of atherosclerosis. Diab Vasc Dis Res 2019; 16:562-576. [PMID: 31530180 PMCID: PMC6787765 DOI: 10.1177/1479164119874469] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Diabetes contributes directly to the development of cardiovascular aortic valve disease. There is currently no drug therapy available for a dysfunctional valve and this urges the need for additional research to identify distinctive mechanisms of cardiovascular aortic valve disease evolution. The aim of this study was to evaluate changes of valvular aortic lesions induced in a hyperlipemic ApoE-/- mouse model by early type 1 diabetes onset (at 4 and 7 days after streptozotocin induction). The haemodynamic valve parameters were evaluated by echography and blood samples and aortic valves were collected. Plasma parameters were measured, and inflammatory, remodelling and osteogenic markers were evaluated in the aortic valves. Next, correlations between all parameters were determined. The results showed early aortic valve dysfunction detected by echography after 1 week of diabetes; lesions were found in the aortic root. Moreover, increased expression of cell adhesion molecules, extracellular matrix remodelling and osteogenic markers were detected in hyperlipemic ApoE-/- diabetic mice. Significant correlations were found between tissue valve biomarkers and plasmatic and haemodynamic parameters. Our study may help to understand the mechanisms of aortic valve disease in the diabetic milieu in order to discover and validate new biomarkers of cardiovascular aortic valve disease in diabetes and reveal new possible targets for nanobiotherapies.
Collapse
Affiliation(s)
| | - Alexandru Filippi
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
| | - Nicoleta Alexandru
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
| | | | - Letitia Ciortan
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
| | - Razvan Macarie
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
| | - Mihaela Vadana
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
| | - Geanina Voicu
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
| | - Sabina Frunza
- Internal Medicine Clinic, Emergency
Clinical Hospital, Bucharest, Romania
| | - Dan Nistor
- Clinical Department of Internal
Medicine, University of Medicine and Pharmacy of Targu Mures, Targu Mures,
Romania
| | - Agneta Simionescu
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
- Clemson University, Clemson, SC,
USA
| | | | - Adriana Georgescu
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
| | - Ileana Manduteanu
- Institute of Cellular Biology and
Pathology ‘Nicolae Simionescu’, Bucharest, Romania
| |
Collapse
|
187
|
Abstract
Unstable coronary plaques that are prone to erosion and rupture are the major cause of acute coronary syndromes. Our expanding understanding of the biological mechanisms of coronary atherosclerosis and rapid technological advances in the field of medical imaging has established cardiac computed tomography as a first-line diagnostic test in the assessment of suspected coronary artery disease, and as a powerful method of detecting the vulnerable plaque and patient. Cardiac computed tomography can provide a noninvasive, yet comprehensive, qualitative and quantitative assessment of coronary plaque burden, detect distinct high-risk morphological plaque features, assess the hemodynamic significance of coronary lesions and quantify the coronary inflammatory burden by tracking the effects of arterial inflammation on the composition of the adjacent perivascular fat. Furthermore, advances in machine learning, computational fluid dynamic modeling, and the development of targeted contrast agents continue to expand the capabilities of cardiac computed tomography imaging. In our Review, we discuss the current role of cardiac computed tomography in the assessment of coronary atherosclerosis, highlighting its dual function as a clinical and research tool that provides a wealth of structural and functional information, with far-reaching diagnostic and prognostic implications.
Collapse
Affiliation(s)
- Evangelos K. Oikonomou
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Henry W. West
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Charalambos Antoniades
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| |
Collapse
|
188
|
Smith ER, Hewitson TD, Holt SG. Diagnostic Tests for Vascular Calcification. Adv Chronic Kidney Dis 2019; 26:445-463. [PMID: 31831123 DOI: 10.1053/j.ackd.2019.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/22/2019] [Accepted: 07/28/2019] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is the heterogeneous endpoint of multiple vascular insults, which varies by arterial bed, the layer of the arterial wall affected, and is propagated by diverse cellular and biochemical mechanisms. A variety of in vivo and ex vivo techniques have been applied to the analysis of VC in preclinical studies, but clinical examination has principally relied on a number of noninvasive and invasive imaging modalities for detection and quantitation. Most imaging methods suffer from suboptimal spatial resolution, leading to the inability to distinguish medial from intimal VC and insufficient sensitivity to detect microcalcifications that are indicative of active mineral deposition and of vulnerable plaques which may be prone to rupture. Serum biomarkers lack specificity for VC and cannot discriminate pathology. Overall, uncertainties surrounding the sensitivity and specificity of different VC testing modalities, the absence of a clear cause-effect relationship, and lack of any evidence-based diagnostic or therapeutic protocols in relation to VC testing in chronic kidney disease has yielded weak or ungraded recommendations for their use in clinical practice. While VC is recognized as a key manifestation of chronic kidney disease-mineral and bone disorder and those with an increasing burden of VC are considered to be at higher cardiovascular risk, routine screening is not currently recommended.
Collapse
|
189
|
Malhotra R, Mauer AC, Lino Cardenas CL, Guo X, Yao J, Zhang X, Wunderer F, Smith AV, Wong Q, Pechlivanis S, Hwang SJ, Wang J, Lu L, Nicholson CJ, Shelton G, Buswell MD, Barnes HJ, Sigurslid HH, Slocum C, Rourke CO, Rhee DK, Bagchi A, Nigwekar SU, Buys ES, Campbell CY, Harris T, Budoff M, Criqui MH, Rotter JI, Johnson AD, Song C, Franceschini N, Debette S, Hoffmann U, Kälsch H, Nöthen MM, Sigurdsson S, Freedman BI, Bowden DW, Jöckel KH, Moebus S, Erbel R, Feitosa MF, Gudnason V, Thanassoulis G, Zapol WM, Lindsay ME, Bloch DB, Post WS, O'Donnell CJ. HDAC9 is implicated in atherosclerotic aortic calcification and affects vascular smooth muscle cell phenotype. Nat Genet 2019; 51:1580-1587. [PMID: 31659325 PMCID: PMC6858575 DOI: 10.1038/s41588-019-0514-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/16/2019] [Indexed: 01/16/2023]
Abstract
Aortic calcification is an important independent predictor of future cardiovascular events. We performed a genome-wide association meta-analysis to determine SNPs associated with the extent of abdominal aortic calcification (n = 9,417) or descending thoracic aortic calcification (n = 8,422). Two genetic loci, HDAC9 and RAP1GAP, were associated with abdominal aortic calcification at a genome-wide level (P < 5.0 × 10-8). No SNPs were associated with thoracic aortic calcification at the genome-wide threshold. Increased expression of HDAC9 in human aortic smooth muscle cells promoted calcification and reduced contractility, while inhibition of HDAC9 in human aortic smooth muscle cells inhibited calcification and enhanced cell contractility. In matrix Gla protein-deficient mice, a model of human vascular calcification, mice lacking HDAC9 had a 40% reduction in aortic calcification and improved survival. This translational genomic study identifies the first genetic risk locus associated with calcification of the abdominal aorta and describes a previously unknown role for HDAC9 in the development of vascular calcification.
Collapse
Affiliation(s)
- Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Andreas C Mauer
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Christian L Lino Cardenas
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiaoling Zhang
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Department of Medicine (Biomedical Genetics Section), Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Florian Wunderer
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Albert V Smith
- Icelandic Heart Association, Kópavogur, Iceland
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Quenna Wong
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Sonali Pechlivanis
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Division of Intramural Research, Bethesda, MD, USA
| | - Judy Wang
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lingyi Lu
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christopher J Nicholson
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Georgia Shelton
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary D Buswell
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hanna J Barnes
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Haakon H Sigurslid
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Charles Slocum
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Caitlin O' Rourke
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - David K Rhee
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Aranya Bagchi
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sagar U Nigwekar
- Harvard Medical School, Boston, MA, USA
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Emmanuel S Buys
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Matthew Budoff
- Division of Cardiology, Department of Medicine and Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Michael H Criqui
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Andrew D Johnson
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Division of Intramural Research, Bethesda, MD, USA
| | - Ci Song
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Division of Intramural Research, Bethesda, MD, USA
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Stephanie Debette
- Inserm U1219, University of Bordeaux, Bordeaux, France
- Department of Neurology, University Hospital of Bordeaux, Bordeaux, France
| | - Udo Hoffmann
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Hagen Kälsch
- Department of Cardiology, Alfried Krupp Hospital, Essen, Germany
- Witten/Herdecke University, Witten, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain GmbH, University of Bonn, Bonn, Germany
| | | | | | | | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
- Centre for Urban Epidemiology, University Hospital Essen, Essen, Germany
| | - Raimund Erbel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, Essen, Germany
| | - Mary F Feitosa
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kópavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - George Thanassoulis
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Warren M Zapol
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mark E Lindsay
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Donald B Bloch
- Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Rheumatology, Allergy, and Immunology; Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher J O'Donnell
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA.
- U.S. Department of Veterans Affairs, Boston, MA, USA.
| |
Collapse
|
190
|
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
|
191
|
Tavakoli S, Sadeghi MM. 18F-Sodium Fluoride Positron Emission Tomography and Plaque Calcification. Circ Cardiovasc Imaging 2019; 12:e008712. [PMID: 30642218 DOI: 10.1161/circimaging.118.008712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sina Tavakoli
- Department of Radiology and Department of Medicine (Vascular Medicine Institute), University of Pittsburgh, PA (S.T.)
| | - Mehran M Sadeghi
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (M.M.S.).,VA Connecticut Healthcare System, West Haven (M.M.S.)
| |
Collapse
|
192
|
Iwai T, Kataoka Y, Otsuka F, Asaumi Y, Nicholls SJ, Noguchi T, Yasuda S. Chronic kidney disease and coronary atherosclerosis: evidences from intravascular imaging. Expert Rev Cardiovasc Ther 2019; 17:707-716. [DOI: 10.1080/14779072.2019.1676150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Takamasa Iwai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yu Kataoka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Fumiyuki Otsuka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yasuhide Asaumi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | | | - Teruo Noguchi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| |
Collapse
|
193
|
Harman JL, Jørgensen HF. The role of smooth muscle cells in plaque stability: Therapeutic targeting potential. Br J Pharmacol 2019; 176:3741-3753. [PMID: 31254285 PMCID: PMC6780045 DOI: 10.1111/bph.14779] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 02/02/2023] Open
Abstract
Events responsible for cardiovascular mortality and morbidity are predominantly caused by rupture of "vulnerable" atherosclerotic lesions. Vascular smooth muscle cells (VSMCs) play a key role in atherogenesis and have historically been considered beneficial for plaque stability. VSMCs constitute the main cellular component of the protective fibrous cap within lesions and are responsible for synthesising strength-giving extracellular matrix components. However, lineage-tracing experiments in mouse models of atherosclerosis have shown that, in addition to the fibrous cap, VSMCs also give rise to many of the cell types found within the plaque core. In particular, VSMCs generate a substantial fraction of lipid-laden foam cells, and VSMC-derived cells expressing markers of macrophages, osteochondrocyte, and mesenchymal stem cells have been observed within lesions. Here, we review recent studies that have changed our perspective on VSMC function in atherosclerosis and discuss how VSMCs could be targeted to increase plaque stability.
Collapse
|
194
|
Messerli AW, Ziada KM, Whayne TF. Dark Spots in Bright Clouds: The Ominous Significance of Spotty Calcification. Angiology 2019; 70:793-794. [DOI: 10.1177/0003319718822651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Adrian W. Messerli
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY, USA
| | - Khaled M. Ziada
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY, USA
| | - Thomas F. Whayne
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
195
|
Association between carotid 18F-NaF and 18F-FDG uptake on PET/CT with ischemic vascular brain disease on MRI in patients with carotid artery disease. Ann Nucl Med 2019; 33:907-915. [PMID: 31571042 DOI: 10.1007/s12149-019-01403-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/24/2019] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Atherosclerosis is a dynamic and complex process characterized by the formation and progression of plaque mediated by various pathophysiologic steps including inflammation and calcification. The present study aimed to evaluate the association between carotid 18F-sodium fluoride (NaF) and 18F-fluorodeoxyglucose (FDG) uptake with the severity of ischemic vascular brain disease on MRI in patients with carotid artery disease. METHODS A total of 28 patients who were scheduled to undergo clinically indicated carotid endarterectomy or stenting for carotid artery disease were examined with 18F-NaF and 18F-FDG PET/CT and brain MRI. The PET/CT images were evaluated by qualitative and semiquantitative analyses. The maximum standardized uptake value (SUV) for the plaque and the average of mean SUV within the lumen of both internal jugular veins was calculated, and the target-to-blood pool ratio (TBR) was determined. The ischemic vascular brain disease on MRI was graded separately in the bilateral hemisphere as 0, 1, 2, and 3, with 0 being absent and 3 being the most severe. RESULTS In two patients, only a unilateral carotid artery was analyzed because of previous indwelling stent. 18F-NaF focal uptake was observed in 50 carotid arteries. 18F-FDG focal uptake was observed in 47 carotid arteries. The mean (± SD) 18F-NaF TBR (2.93 ± 0.89) was significantly higher than the mean (± SD) 18F-FDG TBR (2.41 ± 0.84) (p < 0.001). The mean (± SD) values of 18F-NaF TBR were 2.63 ± 0.76 in grade 1, 2.90 ± 0.91 in grade 2, and 3.81 ± 0.60 in grade 3. Significant differences in 18F-NaF TBR were observed between grades 1 and 3 (p < 0.001) and grades 2 and 3 (p = 0.02). The mean (± SD) values of 18F-FDG TBR were 2.35 ± 0.77 in grade 1, 2.23 ± 0.48 in grade 2, and 2.87 ± 1.32 in grade 3. No significant differences in 18F-FDG TBR were noted between any of the ischemic vascular brain disease grades. CONCLUSIONS These preliminary results suggest that carotid 18F-NaF uptake in patients with carotid artery disease may be associated with the severity of the ischemic vascular brain disease observed on MRI.
Collapse
|
196
|
Vancheri F, Longo G, Vancheri S, Danial JSH, Henein MY. Coronary Artery Microcalcification: Imaging and Clinical Implications. Diagnostics (Basel) 2019; 9:E125. [PMID: 31547506 PMCID: PMC6963848 DOI: 10.3390/diagnostics9040125] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 02/06/2023] Open
Abstract
Strategies to prevent acute coronary and cerebrovascular events are based on accurate identification of patients at increased cardiovascular (CV) risk who may benefit from intensive preventive measures. The majority of acute CV events are precipitated by the rupture of the thin cap overlying the necrotic core of an atherosclerotic plaque. Hence, identification of vulnerable coronary lesions is essential for CV prevention. Atherosclerosis is a highly dynamic process involving cell migration, apoptosis, inflammation, osteogenesis, and intimal calcification, progressing from early lesions to advanced plaques. Coronary artery calcification (CAC) is a marker of coronary atherosclerosis, correlates with clinically significant coronary artery disease (CAD), predicts future CV events and improves the risk prediction of conventional risk factors. The relative importance of coronary calcification, whether it has a protective effect as a stabilizing force of high-risk atherosclerotic plaque has been debated until recently. The extent of calcium in coronary arteries has different clinical implications. Extensive plaque calcification is often a feature of advanced and stable atherosclerosis, which only rarely results in rupture. These macroscopic vascular calcifications can be detected by computed tomography (CT). The resulting CAC scoring, although a good marker of overall coronary plaque burden, is not useful to identify vulnerable lesions prone to rupture. Unlike macrocalcifications, spotty microcalcifications assessed by intravascular ultrasound or optical coherence tomography strongly correlate with plaque instability. However, they are below the resolution of CT due to limited spatial resolution. Microcalcifications develop in the earliest stages of coronary intimal calcification and directly contribute to plaque rupture producing local mechanical stress on the plaque surface. They result from a healing response to intense local macrophage inflammatory activity. Most of them show a progressive calcification transforming the early stage high-risk microcalcification into the stable end-stage macroscopic calcification. In recent years, new developments in noninvasive cardiovascular imaging technology have shifted the study of vulnerable plaques from morphology to the assessment of disease activity of the atherosclerotic lesions. Increased disease activity, detected by positron emission tomography (PET) and magnetic resonance (MR), has been shown to be associated with more microcalcification, larger necrotic core and greater rates of events. In this context, the paradox of increased coronary artery calcification observed in statin trials, despite reduced CV events, can be explained by the reduction of coronary inflammation induced by statin which results in more stable macrocalcification.
Collapse
Affiliation(s)
| | - Giovanni Longo
- Cardiovascular and Interventional Department, S.Elia Hospital, 93100 Caltanissetta, Italy.
| | - Sergio Vancheri
- Radiology Department, I.R.C.C.S. Policlinico San Matteo, 27100 Pavia, Italy.
| | - John S H Danial
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK.
| | - Michael Y Henein
- Institute of Public Health and Clinical Medicine, Umea University, 901 87 Umea, Sweden.
- Institute of Environment & Health and Societies, Brunel University, Middlesex SW17 0RE, UK.
- Molecular and Clinical Sciences Research Institute, St George's University, London UB8 3PH, UK.
| |
Collapse
|
197
|
Zabirnyk A, Ferrer MD, Bogdanova M, Pérez MM, Salcedo C, Kaljusto ML, Kvitting JPE, Stensløkken KO, Perelló J, Vaage J. SNF472, a novel anti-crystallization agent, inhibits induced calcification in an in vitro model of human aortic valve calcification. Vascul Pharmacol 2019; 122-123:106583. [PMID: 31437530 DOI: 10.1016/j.vph.2019.106583] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/03/2019] [Accepted: 08/18/2019] [Indexed: 12/27/2022]
Abstract
The purpose of the present study was to investigate whether SNF472, the hexasodium salt of myo-inositol hexaphosphate (IP6 or phytate): 1. Inhibits induced calcification in cultured aortic valve interstitial cells (VIC) as an in vitro model of aortic valve stenosis and 2. Whether inhibition is different in VIC obtained from healthy and calcified aortic valves. VIC from healthy (n = 5) and calcified (n = 7) human aortic valves were seeded in basic growth medium, osteogenic differentiation medium alone, or in osteogenic medium with SNF472 (3, 10, and 30 μM) and cultivated for 3 weeks. Calcification was quantified spectrophotometrically after Alizarin Red staining. In VIC from calcified valves, a complete inhibition of calcification was observed with SNF472 concentrations of 10 and 30 μM (p < .01), significantly stronger than in VIC from healthy valves. When SNF472 was added to VIC after 1 week in osteogenic medium, 30 and 100 μM SNF472 inhibited the progression of ongoing calcification by 81 and 100% (p < .01), respectively. The same concentrations of SNF472 given after 2 weeks reduced calcification by 35 and 40% respectively (not significant). SNF472 inhibited both the formation and the progression of calcification with the strongest effect in VIC from calcified valves.
Collapse
Affiliation(s)
- A Zabirnyk
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - M D Ferrer
- Sanifit Therapeutics, Palma, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands, Palma, Spain
| | - M Bogdanova
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | - M-L Kaljusto
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - J-P E Kvitting
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - K-O Stensløkken
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - J Perelló
- Sanifit Therapeutics, Palma, Spain; Department of Chemistry, University of the Balearic Islands, Palma, Spain
| | - J Vaage
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
198
|
Strauss HW, Nakahara T, Narula N, Narula J. Vascular Calcification: The Evolving Relationship of Vascular Calcification to Major Acute Coronary Events. J Nucl Med 2019; 60:1207-1212. [PMID: 31350320 DOI: 10.2967/jnumed.119.230276] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022] Open
Abstract
Calcification in a coronary artery is accepted as definite evidence of coronary atherosclerosis. The extent and density of calcification, as combined in the Agatston score, is associated with the risk of a patient experiencing a major acute coronary event. Atherosclerosis occurs because damaged endothelial cells allow low-density lipoprotein cholesterol (LDLc) to leak into subintimal tissue. Proteoglycans in subendothelial collagen have a high affinity for LDLc, retaining the lipoprotein cholesterol complex. As the endothelial damage is repaired, the subintimal LDLc is trapped. Retained LDLc induces an inflammatory response in the overlying endothelium, causing the endothelium to express chemotactic peptides. Chemotactic peptides attract circulating monocytes, which follow the concentration gradient, enter the tissue, and become tissue macrophages to phagocytize and digest the irritating LDLc in the atheroma. In the process of digesting LDLc, enzymes in the macrophages oxidize the LDLc complex. Oxidized LDL is toxic to macrophages; when present in sufficient quantity, it may cause death of macrophages, contributing to inflammation in the atheroma. In a necrotic inflammatory lesion, the regulatory mechanisms that control tissue concentrations of calcium and phosphorus are lost, allowing the solubility product of calcium phosphate to be exceeded, resulting in the formation of microscopic calcium-phosphate crystals. With ongoing inflammation, additional calcium-phosphate crystals are formed, which may aggregate. When these aggregated calcium phosphate crystals exceed 1 mm, the lesions become visible on clinical CT as coronary calcifications. Serial gated CT scans of the heart have demonstrated that once formed, CT-visible calcifications do not decrease significantly in size but may increase.
Collapse
Affiliation(s)
- H William Strauss
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Takehiro Nakahara
- Department of Diagnostic Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Navneet Narula
- Department of Pathology, New York University School of Medicine, New York, New York; and
| | - Jagat Narula
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
199
|
Durham AL, Speer MY, Scatena M, Giachelli CM, Shanahan CM. Role of smooth muscle cells in vascular calcification: implications in atherosclerosis and arterial stiffness. Cardiovasc Res 2019. [PMID: 29514202 PMCID: PMC5852633 DOI: 10.1093/cvr/cvy010] [Citation(s) in RCA: 726] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Vascular calcification is associated with a significant increase in all-cause mortality and atherosclerotic plaque rupture. Calcification has been determined to be an active process driven in part by vascular smooth muscle cell (VSMC) transdifferentiation within the vascular wall. Historically, VSMC phenotype switching has been viewed as binary, with the cells able to adopt a physiological contractile phenotype or an alternate ‘synthetic’ phenotype in response to injury. More recent work, including lineage tracing has however revealed that VSMCs are able to adopt a number of phenotypes, including calcific (osteogenic, chondrocytic, and osteoclastic), adipogenic, and macrophagic phenotypes. Whilst the mechanisms that drive VSMC differentiation are still being elucidated it is becoming clear that medial calcification may differ in several ways from the intimal calcification seen in atherosclerotic lesions, including risk factors and specific drivers for VSMC phenotype changes and calcification. This article aims to compare and contrast the role of VSMCs in driving calcification in both atherosclerosis and in the vessel media focusing on the major drivers of calcification, including aging, uraemia, mechanical stress, oxidative stress, and inflammation. The review also discusses novel findings that have also brought attention to specific pro- and anti-calcifying proteins, extracellular vesicles, mitochondrial dysfunction, and a uraemic milieu as major determinants of vascular calcification.
Collapse
Affiliation(s)
- Andrew L Durham
- Division of Cardiology, James Black Centre, Kings College London, Denmark Hill, London, SE5 9NU, UK
| | - Mei Y Speer
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Marta Scatena
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Cecilia M Giachelli
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Catherine M Shanahan
- Division of Cardiology, James Black Centre, Kings College London, Denmark Hill, London, SE5 9NU, UK
| |
Collapse
|
200
|
|