1
|
Kiełbowski K, Skórka P, Plewa P, Bakinowska E, Pawlik A. The Role of Alarmins in the Pathogenesis of Atherosclerosis and Myocardial Infarction. Curr Issues Mol Biol 2024; 46:8995-9015. [PMID: 39194749 DOI: 10.3390/cimb46080532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Atherosclerosis is a condition that is associated with lipid accumulation in the arterial intima. Consequently, the enlarging lesion, which is also known as an atherosclerotic plaque, may close the blood vessel lumen, thus leading to organ ischaemia. Furthermore, the plaque may rupture and initiate the formation of a thrombus, which can cause acute ischaemia. Atherosclerosis is a background pathological condition that can eventually lead to major cardiovascular diseases such as acute coronary syndrome or ischaemic stroke. The disorder is associated with an altered profile of alarmins, stress response molecules that are secreted due to cell injury or death and that induce inflammatory responses. High-mobility group box 1 (HMGB1), S100 proteins, interleukin-33, and heat shock proteins (HSPs) also affect the behaviour of endothelial cells and vascular smooth muscle cells (VSMCs). Thus, alarmins control the inflammatory responses of endothelial cells and proliferation of VSMCs, two important processes implicated in the pathogenesis of atherosclerosis. In this review, we will discuss the role of alarmins in the pathophysiology of atherosclerosis and myocardial infarction.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Patryk Skórka
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Paulina Plewa
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
2
|
Li W, Su SA, Chen J, Shen Y, Ma H, Xiang M. EphrinB2 drives osteogenic fate of adult cardiac fibroblasts in a calcium influx dependent manner. Am J Physiol Cell Physiol 2023; 325:C69-C78. [PMID: 37212547 DOI: 10.1152/ajpcell.00301.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023]
Abstract
Cardiac calcification is a crucial but underrecognized pathological process, greatly increasing the risk of cardiovascular diseases. Little is known about how cardiac fibroblasts, as a central mediator, facilitate abnormal mineralization. Erythropoietin-producing hepatoma interactor B2 (EphrinB2), previously identified as an angiogenic regulator, is involved in fibroblast activation, while its role in the osteogenic differentiation of cardiac fibroblasts is unknown. Bioinformatics analysis was conducted to characterize the expression of the Ephrin family in human calcified aortic valves and calcific mouse hearts. The effects of EphrinB2 on cardiac fibroblasts to adopt osteogenic fate was determined by gain- and loss-of-function. EphrinB2 mRNA level was downregulated in calcified aortic valves and mouse hearts. Knockdown of EphrinB2 attenuated mineral deposits in adult cardiac fibroblasts, whereas overexpression of EphrinB2 promoted their osteogenic differentiation. RNA sequencing data implied that Ca2+-related S100/receptor for advanced glycation end products (RAGE) signaling may mediate EphrinB2-induced mineralization in cardiac fibroblasts. Moreover, L-type calcium channel blockers inhibited osteogenic differentiation of cardiac fibroblasts, implying a critical role in Ca2+ influx. In conclusion, our data illustrated an unrecognized role of EphrinB2, which functions as a novel osteogenic regulator in the heart through Ca2+ signaling and could be a potential therapeutic target in cardiovascular calcification.NEW & NOTEWORTHY In this study, we observed that adult cardiac fibroblasts but not neonatal cardiac fibroblasts exhibit the ability of osteogenic differentiation. EphrinB2 promoted osteogenic differentiation of cardiac fibroblasts through activating Ca2+-related S100/RAGE signaling. Inhibition of Ca2+ influx using L-type calcium channel blockers inhibited EphrinB2-mediated calcification of cardiac fibroblasts. Our data implied an unrecognized role of EphrinB2 in regulating cardiac calcification though Ca2+-related signaling, suggesting a potential therapeutic target of cardiovascular calcification.
Collapse
Affiliation(s)
- Wudi Li
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Sheng-An Su
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jian Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Yimin Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
3
|
Zhou Y, Zha Y, Yang Y, Ma T, Li H, Liang J. S100 proteins in cardiovascular diseases. Mol Med 2023; 29:68. [PMID: 37217870 DOI: 10.1186/s10020-023-00662-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
Cardiovascular diseases have become a serious threat to human health and life worldwide and have the highest fatality rate. Therefore, the prevention and treatment of cardiovascular diseases have become a focus for public health experts. The expression of S100 proteins is cell- and tissue-specific; they are implicated in cardiovascular, neurodegenerative, and inflammatory diseases and cancer. This review article discusses the progress in the research on the role of S100 protein family members in cardiovascular diseases. Understanding the mechanisms by which these proteins exert their biological function may provide novel concepts for preventing, treating, and predicting cardiovascular diseases.
Collapse
Affiliation(s)
- Yue Zhou
- Medical College, Yangzhou University, Yangzhou, China
| | - Yiwen Zha
- Medical College, Yangzhou University, Yangzhou, China
| | - Yongqi Yang
- Medical College, Yangzhou University, Yangzhou, China
| | - Tan Ma
- Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Hongliang Li
- Medical College, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
| | - Jingyan Liang
- Medical College, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China.
| |
Collapse
|
4
|
Woo SH, Kim DY, Choi JH. Roles of Vascular Smooth Muscle Cells in Atherosclerotic Calcification. J Lipid Atheroscler 2023; 12:106-118. [PMID: 37265849 PMCID: PMC10232217 DOI: 10.12997/jla.2023.12.2.106] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/08/2023] [Accepted: 03/17/2023] [Indexed: 06/03/2023] Open
Abstract
The accumulation of calcium in atherosclerotic plaques is a prominent feature of advanced atherosclerosis, and it has a strong positive correlation with the total burden of atherosclerosis. Atherosclerotic calcification usually appears first at the necrotic core, indicating that cell death and inflammatory processes are involved in calcification. During atherosclerotic inflammation, various cell types, such as vascular smooth muscle cells, nascent resident pericytes, circulating stem cells, or adventitial cells, have been assumed to differentiate into osteoblastic cells, which lead to vascular calcification. Among these cell types, vascular smooth muscle cells are considered a major contributor to osteochondrogenic cells in the atherosclerotic milieu. In this review, we summarize the molecular mechanisms underlying the osteochondrogenic switch of vascular smooth muscle cells in atherosclerotic plaques.
Collapse
Affiliation(s)
- Sang-Ho Woo
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute of Natural Sciences, Research Institute for Convergence of Basic Sciences, Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| |
Collapse
|
5
|
The Role of Hydrogen Sulfide in Plaque Stability. Antioxidants (Basel) 2022; 11:antiox11122356. [PMID: 36552564 PMCID: PMC9774534 DOI: 10.3390/antiox11122356] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Atherosclerosis is the greatest contributor to cardiovascular events and is involved in the majority of deaths worldwide. Plaque rapture or erosion precipitates life-threatening thrombi, resulting in the obstruction blood flow to the heart (acute coronary syndrome), brain (ischemic stroke) or low extremities (peripheral vascular diseases). Among these events, major causation dues to the plaque rupture. Although the initiation, procession, and precise time of controlling plaque rupture are unclear, foam cell formation and apoptosis, cell death, extracellular matrix components, protease expression and activity, local inflammation, intraplaque hemorrhage, and calcification contribute to the plaque instability. These alterations tightly associate with the function regulation of intraplaque various cell populations. Hydrogen sulfide (H2S) is gasotransmitter derived from methionine metabolism and exerts a protective role in the genesis of atherosclerosis. Recent progress also showed H2S mediated the plaque stability. In this review, we discuss the progress of endogenous H2S modulation on functions of vascular smooth muscle cells, monocytes/macrophages, and T cells, and the molecular mechanism in plaque stability.
Collapse
|
6
|
Inciarte-Mundo J, Frade-Sosa B, Sanmartí R. From bench to bedside: Calprotectin (S100A8/S100A9) as a biomarker in rheumatoid arthritis. Front Immunol 2022; 13:1001025. [PMID: 36405711 PMCID: PMC9672845 DOI: 10.3389/fimmu.2022.1001025] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/19/2022] [Indexed: 12/30/2022] Open
Abstract
S100A9/S100A8 (calprotectin), a member of the S100 protein family, has been shown to play a pivotal role in innate immunity activation. Calprotectin plays a critical role in the pathogenesis of rheumatoid arthritis (RA), as it triggers chemotaxis, phagocyte migration and modulation of neutrophils and macrophages. Higher calprotectin levels have been found in synovial fluid, plasma, and serum from RA patients. Recent studies have demonstrated better correlations between serum or plasma calprotectin and composite inflammatory disease activity indexes than c-reactive protein (CRP) or the erythrocyte sedimentation rate (ESR). Calprotectin serum levels decreased after treatment, independently of the DMARD type or strategy. Calprotectin has shown the strongest correlations with other sensitive techniques to detect inflammation, such as ultrasound. Calprotectin independently predicts radiographic progression. However, its value as a biomarker of treatment response and flare after tapering is unclear. This update reviews the current understanding of calprotectin in RA and discusses possible applications as a biomarker in clinical practice.
Collapse
Affiliation(s)
- José Inciarte-Mundo
- Biological aggression and Response Mechanisms, Inflammatory joint diseases (IJDs), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Beatriz Frade-Sosa
- Rheumatology Department, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Raimon Sanmartí
- Biological aggression and Response Mechanisms, Inflammatory joint diseases (IJDs), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain,Rheumatology Department, Hospital Clinic, University of Barcelona, Barcelona, Spain,*Correspondence: Raimon Sanmartí,
| |
Collapse
|
7
|
Jeong JH, Lee DH, Song J. HMGB1 signaling pathway in diabetes-related dementia: Blood-brain barrier breakdown, brain insulin resistance, and Aβ accumulation. Biomed Pharmacother 2022; 150:112933. [PMID: 35413600 DOI: 10.1016/j.biopha.2022.112933] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/28/2022] Open
Abstract
Diabetes contributes to the onset of various diseases, including cancer and cardiovascular and neurodegenerative diseases. Recent studies have highlighted the similarities and relationship between diabetes and dementia as an important issue for treating diabetes-related cognitive deficits. Diabetes-related dementia exhibits several features, including blood-brain barrier disruption, brain insulin resistance, and Aβ over-accumulation. High-mobility group box1 (HMGB1) is a protein known to regulate gene transcription and cellular mechanisms by binding to DNA or chromatin via receptor for advanced glycation end-products (RAGE) and toll-like receptor 4 (TLR4). Recent studies have demonstrated that the interplay between HMGB1, RAGE, and TLR4 can impact both neuropathology and diabetic alterations. Herein, we review the recent research regarding the roles of HMGB1-RAGE-TLR4 axis in diabetes-related dementia from several perspectives and emphasize the importance of the influence of HMGB1 in diabetes-related dementia.
Collapse
Affiliation(s)
- Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| | - Dong Hoon Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School, and Chonnam National University Hwasun Hospital, Hwasun 58128, Jeollanam-do, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| |
Collapse
|
8
|
Xiang Q, Tian F, Xu J, Du X, Zhang S, Liu L. New insight into dyslipidemia‐induced cellular senescence in atherosclerosis. Biol Rev Camb Philos Soc 2022; 97:1844-1867. [PMID: 35569818 PMCID: PMC9541442 DOI: 10.1111/brv.12866] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/18/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022]
Abstract
Atherosclerosis, characterized by lipid‐rich plaques in the arterial wall, is an age‐related disorder and a leading cause of mortality worldwide. However, the specific mechanisms remain complex. Recently, emerging evidence has demonstrated that senescence of various types of cells, such as endothelial cells (ECs), vascular smooth muscle cells (VSMCs), macrophages, endothelial progenitor cells (EPCs), and adipose‐derived mesenchymal stem cells (AMSCs) contributes to atherosclerosis. Cellular senescence and atherosclerosis share various causative stimuli, in which dyslipidemia has attracted much attention. Dyslipidemia, mainly referred to elevated plasma levels of atherogenic lipids or lipoproteins, or functional impairment of anti‐atherogenic lipids or lipoproteins, plays a pivotal role both in cellular senescence and atherosclerosis. In this review, we summarize the current evidence for dyslipidemia‐induced cellular senescence during atherosclerosis, with a focus on low‐density lipoprotein (LDL) and its modifications, hydrolysate of triglyceride‐rich lipoproteins (TRLs), and high‐density lipoprotein (HDL), respectively. Furthermore, we describe the underlying mechanisms linking dyslipidemia‐induced cellular senescence and atherosclerosis. Finally, we discuss the senescence‐related therapeutic strategies for atherosclerosis, with special attention given to the anti‐atherosclerotic effects of promising geroprotectors as well as anti‐senescence effects of current lipid‐lowering drugs.
Collapse
Affiliation(s)
- Qunyan Xiang
- Department of Geriatrics, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Institute of Aging and Age‐related Disease Research Central South University Changsha Hunan 410011 PR China
| | - Feng Tian
- Department of Geriatric Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450000 PR China
| | - Jin Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| | - Xiao Du
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| | - Shilan Zhang
- Department of Gastroenterology, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| |
Collapse
|
9
|
Jeong J, Cho S, Lee BS, Seo M, Jang Y, Lim S, Park S. Soluble RAGE attenuates Ang II-induced arterial calcification via inhibiting AT1R-HMGB1-RAGE axis. Atherosclerosis 2022; 346:53-62. [DOI: 10.1016/j.atherosclerosis.2022.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/30/2022]
|
10
|
Mao L, Yin R, Yang L, Zhao D. Role of advanced glycation end products on vascular smooth muscle cells under diabetic atherosclerosis. Front Endocrinol (Lausanne) 2022; 13:983723. [PMID: 36120471 PMCID: PMC9470882 DOI: 10.3389/fendo.2022.983723] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease and leading cause of cardiovascular diseases. The progression of AS is a multi-step process leading to high morbidity and mortality. Hyperglycemia, dyslipidemia, advanced glycation end products (AGEs), inflammation and insulin resistance which strictly involved in diabetes are closely related to the pathogenesis of AS. A growing number of studies have linked AGEs to AS. As one of the risk factors of cardiac metabolic diseases, dysfunction of VSMCs plays an important role in AS pathogenesis. AGEs are increased in diabetes, participate in the occurrence and progression of AS through multiple molecular mechanisms of vascular cell injury. As the main functional cells of vascular, vascular smooth muscle cells (VSMCs) play different roles in each stage of atherosclerotic lesions. The interaction between AGEs and receptor for AGEs (RAGE) accelerates AS by affecting the proliferation and migration of VSMCs. In addition, increasing researches have reported that AGEs promote osteogenic transformation and macrophage-like transformation of VSMCs, and affect the progression of AS through other aspects such as autophagy and cell cycle. In this review, we summarize the effect of AGEs on VSMCs in atherosclerotic plaque development and progression. We also discuss the AGEs that link AS and diabetes mellitus, including oxidative stress, inflammation, RAGE ligands, small noncoding RNAs.
Collapse
Affiliation(s)
| | | | | | - Dong Zhao
- *Correspondence: Longyan Yang, ; Dong Zhao,
| |
Collapse
|
11
|
Sun XJ, Liu NF. Diabetic mellitus, vascular calcification and hypoxia: A complex and neglected tripartite relationship. Cell Signal 2021; 91:110219. [PMID: 34921978 DOI: 10.1016/j.cellsig.2021.110219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/11/2021] [Accepted: 12/11/2021] [Indexed: 11/15/2022]
Abstract
DM (diabetic mellitus) and its common vascular complications VC (vascular calcification), are increasingly harmful to human health. In recent years, the research on the relationship between DM and VC is also deepening. Hypoxia, as one of the pathogenic factors of many disease models, is also closely related to the occurrence of DM and VC. There are some studies on the role of hypoxia in the pathogenesis of DM and VC respectively, but no one has made an in-depth summary of the systematic connection between hypoxia, DM and VC. Therefore, what we want to review in this article are the relationship between DM, VC and hypoxia, respectively, as well as the role of hypoxia in the development of DM and VC, which has little concern but is a novel and potentially target that may provide some new ideas for the prevention and treatment of DM, VC, especially diabetic VC.
Collapse
Affiliation(s)
- Xue-Jiao Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, PR China.
| |
Collapse
|
12
|
MicroRNA-146a Serves as a Biomarker for Adverse Prognosis of ST-Segment Elevation Myocardial Infarction. Cardiovasc Ther 2021; 2021:2923441. [PMID: 34786024 PMCID: PMC8561321 DOI: 10.1155/2021/2923441] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/09/2021] [Indexed: 11/18/2022] Open
Abstract
Objective This study is aimed at exploring the underlying molecular mechanisms of ST-segment elevation myocardial infarction (STEMI) and provides potential clinical prognostic biomarkers for STEMI. Methods The GSE60993 dataset was downloaded from the GEO database, and the differentially expressed genes (DEGs) between STEMI and control groups were screened. Enrichment analysis of the DEGs was subsequently performed using the DAVID database. A protein-protein interaction network was constructed, and hub genes were identified. The hub genes in patients were then validated by quantitative reverse transcription-PCR. Furthermore, hub gene-miRNA interactions were evaluated using the miRTarBase database. Finally, patient data on classical cardiovascular risk factors were collected, and plasma microRNA-146a (miR-146a) levels were detected. An individualized nomogram was constructed based on multivariate Cox regression analysis. Results A total of 239 DEGs were identified between the STEMI and control groups. Expression of S100A12 and miR-146a was significantly upregulated in STEMI samples compared with controls. STEMI patients with high levels of miR-146a had a higher risk of major adverse cardiovascular events (MACEs) than those with low levels of miR-146a (log-rank P = 0.034). Multivariate Cox regression analysis identified five statistically significant variables, including age, hypertension, diabetes mellitus, white blood cells, and miR-146a. A nomogram was constructed to estimate the likelihood of a MACE at one, two, and three years after STEMI. Conclusion The incidence of MACEs in STEMI patients expressing high levels of miR-146a was significantly greater than in those expressing low levels. MicroRNA-146a can serve as a biomarker for adverse prognosis of STEMI and might function in its pathogenesis by targeting S100A12, which may exert its role via an inflammatory response. In addition, our study presents a valid and practical model to assess the probability of MACEs within three years of STEMI.
Collapse
|
13
|
Jiang X, Huang CM, Feng CM, Xu Z, Fu L, Wang XM. Associations of Serum S100A12 With Severity and Prognosis in Patients With Community-Acquired Pneumonia: A Prospective Cohort Study. Front Immunol 2021; 12:714026. [PMID: 34745092 PMCID: PMC8569229 DOI: 10.3389/fimmu.2021.714026] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022] Open
Abstract
Background Previous studies indicated the calcium-binding protein S100A12 to be involved in the pathophysiology of pulmonary inflammatory diseases. However, the role of S100A12 has remained elusive in patients with community-acquired pneumonia (CAP). Therefore, the purpose of this prospective cohort study was to evaluate the association between serum S100A12 with severity and prognosis in CAP patients. Methods Two groups with either 239 CAP patients or 239 healthy controls were enrolled in our study. Fasting blood and clinical characteristics were collected. On admission, serum S100A12 was measured using enzyme-linked immunosorbent assay (ELISA). Results Serum S100A12 was increased in CAP patients compared to control subjects. Furthermore, serum S100A12 was elevated according to the severity of CAP. Correlative analysis suggested that the level of serum S100A12 was associated with blood routine indices, renal function markers, inflammatory cytokines and other clinical parameters among CAP patients. Additionally, linear and logistical regression analyses indicated that serum S100A12 was positively associated with CAP severity scores in CAP patients. In addition, the association of high serum S100A12 and prognosis was accessed using a follow-up research. Elevated serum S100A12 on admission increased the risk of death and hospital stay in CAP patients during hospitalization. Conclusions Elevated serum S100A12 on admission is positively associated with the severity and adverse prognosis in CAP patients, suggesting that S100A12 may involve in the pathophysiological process of CAP. The titre of serum S100A12 may be used as a biomarker for diagnosis and prognosis among CAP patients.
Collapse
Affiliation(s)
- Xiao Jiang
- Department of Nephrology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chun-Mei Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Chun-Mei Feng
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zheng Xu
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lin Fu
- Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Toxicology, Anhui Medical University, Hefei, China
| | - Xin-Ming Wang
- Department of Pharmacy, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, China
| |
Collapse
|
14
|
Sitagliptin attenuates arterial calcification by downregulating oxidative stress-induced receptor for advanced glycation end products in LDLR knockout mice. Sci Rep 2021; 11:17851. [PMID: 34497344 PMCID: PMC8426400 DOI: 10.1038/s41598-021-97361-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/24/2021] [Indexed: 12/25/2022] Open
Abstract
Diabetes is a complex disease characterized by hyperglycemia, dyslipidemia, and insulin resistance. Plasma advanced glycation end products (AGEs) activated the receptor for advanced glycation end products (RAGE) and the activation of RAGE is implicated to be the pathogenesis of type 2 diabetic mellitus (T2DM) patient vascular complications. Sitagliptin, a dipeptidyl peptidase-4 (DPP4) inhibitor, is a new oral hypoglycemic agent for the treatment of T2DM. However, the beneficial effects on vascular calcification remain unclear. In this study, we used a high-fat diet (HFD)-fed low-density lipoprotein receptor deficiency (LDLR−/−) mice model to investigate the potential effects of sitagliptin on HFD-induced arterial calcification. Mice were randomly divided into 3 groups: (1) normal diet group, (2) HFD group and (3) HFD + sitagliptin group. After 24 weeks treatment, we collected the blood for chemistry parameters and DPP4 activity measurement, and harvested the aorta to evaluate calcification using immunohistochemistry and calcium content. To determine the effects of sitagliptin, tumor necrosis factor (TNF)-α combined with S100A12 was used to induce oxidative stress, activation of nicotinamide adenine dinucleotide phosphate (NADPH), up-regulation of bone markers and RAGE expression, and cell calcium deposition on human aortic smooth muscle cells (HASMCs). We found that sitagliptin effectively blunted the HFD-induced artery calcification and significantly lowered the levels of fasting serum glucose, triglyceride (TG), nitrotyrosine and TNF-α, decreased the calcium deposits, and reduced arterial calcification. In an in-vitro study, both S100A12 and TNF-α stimulated RAGE expression and cellular calcium deposits in HASMCs. The potency of S100A12 on HASMCs was amplified by the presence of TNF-α. Sitagliptin and Apocynin (APO), an NADPH oxidase inhibitor, inhibited the TNF-α + S100A12-induced NADPH oxidase and nuclear factor (NF)-κB activation, cellular oxidative stress, RAGE expression, osteo transcription factors expression and calcium deposition. In addition, treatment with sitagliptin, knockdown of RAGE or TNF-α receptor blunted the TNF-α + S100A12-induced RAGE expression. Our findings suggest that sitagliptin may suppress the initiation and progression of arterial calcification by inhibiting the activation of NADPH oxidase and NF-κB, followed by decreasing the expression of RAGE.
Collapse
|
15
|
Singh H, Rai V, Agrawal DK. Discerning the promising binding sites of S100/calgranulins and their therapeutic potential in atherosclerosis. Expert Opin Ther Pat 2021; 31:1045-1057. [PMID: 34056993 DOI: 10.1080/13543776.2021.1937122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Atherosclerosis is a chronic inflammatory disease in which the members of S100 family proteins (calgranulins) bind with their receptors, particularly receptor for advanced glycation end products (RAGE) and toll-like receptor-4 (TLR-4) and play a key role in the pathogenesis and progression of disease. Thus, these proteins could be considered as potential biomarkers and therapeutic targets in the treatment of atherosclerotic inflammation. AREAS COVERED This review summarizes the pathology of S100A8, S100A9, and S100A12 in the development of atherosclerosis and reveals key structural features of these proteins which are potentially critical in their pathological effects. This article focuses on the translational significance of antagonizing these proteins by using small molecules in patent literature, clinical and preclinical studies and also discusses future approaches that could be employed to block these proteins in the treatment of atherosclerosis. EXPERT OPINION Based on the critical role of S100/calgranulins in the regulation of atherosclerosis, these proteins are potential targets to develop better therapeutic options in the treatment of inflammatory diseases. However, further research is still needed to clarify their exact molecular mechanism by analyzing their detailed structural features that can expedite future research to develop novel therapeutics against these proteins to treat atherosclerotic inflammation.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
16
|
Komatsu T, Ayaori M, Uto-Kondo H, Hayashi K, Tamura K, Sato H, Sasaki M, Nishida T, Takiguchi S, Yakushiji E, Nakaya K, Ikewaki K. Atorvastatin Reduces Circulating S100A12 Levels in Patients with Carotid Atherosclerotic Plaques - A Link with Plaque Inflammation. J Atheroscler Thromb 2021; 29:775-784. [PMID: 33952812 PMCID: PMC9135643 DOI: 10.5551/jat.61630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Aims: Inflammation is involved in various processes of atherosclerosis development. Serum C-reactive protein (CRP) levels, a predictor for cardiovascular risk, are reportedly reduced by statins. However, several studies have demonstrated that CRP is a bystander during atherogenesis. While S100A12 has been focused on as an inflammatory molecule, it remains unclear whether statins affect circulating S100A12 levels. Here, we investigated whether atorvastatin treatment affected S100A12 and which biomarkers were correlated with changes in arterial inflammation.
Methods: We performed a prospective, randomized open-labeled trial on whether atorvastatin affected arterial (carotid and thoracic aorta) inflammation using18fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) and inflammatory markers. Thirty-one statin-naïve patients with carotid atherosclerotic plaques were randomized to either a group receiving dietary management (n=15) or one receiving atorvastatin (10mg/day,n=16) for 12weeks.18F-FDG-PET/CT and flow-mediated vasodilation (FMD) were performed, the latter to evaluate endothelial function.
Results: Atorvastatin, but not the diet-only treatment, significantly reduced LDL-cholesterol (LDL-C, -43%), serum CRP (-37%) and S100A12 levels (-28%) and improved FMD (+38%).18F-FDG-PET/CT demonstrated that atorvastatin, but not the diet-only treatment, significantly reduced accumulation of18F-FDG in the carotid artery and thoracic aorta. A multivariate analysis revealed that reduction in CRP, S100A12, LDL-C, oxidized-LDL, and increase in FMD were significantly associated with reduced arterial inflammation in the thoracic aorta, but not in the carotid artery.
Conclusions: Atorvastatin treatment reduced S100A12/CRP levels, and the changes in these circulating markers mirrored the improvement in arterial inflammation. Our observations suggest that S100A12 may be an emerging therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Tomohiro Komatsu
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College
| | - Makoto Ayaori
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College.,Tokorozawa Heart Center
| | - Harumi Uto-Kondo
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College
| | | | | | - Hiroki Sato
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Makoto Sasaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College
| | - Takafumi Nishida
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College
| | - Shunichi Takiguchi
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College
| | - Emi Yakushiji
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College
| | - Kazuhiro Nakaya
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College
| | - Katsunori Ikewaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College
| |
Collapse
|
17
|
Liang H, Cui Y, Bu H, Liu H, Yan P, Cui L, Chen L. Value of S100A12 in predicting in-stent restenosis in patients with coronary drug-eluting stent implantation. Exp Ther Med 2020; 20:211-218. [PMID: 32536993 PMCID: PMC7282035 DOI: 10.3892/etm.2020.8721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/21/2019] [Indexed: 01/08/2023] Open
Abstract
In-stent restenosis (ISR) after drug-eluting stent (DES) placement has recently emerged as a major concern for cardiologists. Identification of biomarkers to predict ISR may be invaluable for tailored management strategies. The present study aimed to evaluate the prognostic utility of circulating S100 calcium-binding protein A12 (S100A12) for ISR. Out of 2,443 patients with DES-based percutaneous coronary intervention (PCI) and follow-up angiography at ~1 year after DES-based PCI, 258 patients were diagnosed with ISR and 258 patients without ISR were randomly selected as controls. Serum S100A12 levels were determined in the two subsets on admission. The association between ISR and the circulating levels of S100A12 was determined by constructing two multivariate stepwise logistic regression models. In addition, S100A12 was assessed for its ability to predict ISR using receiver operating characteristic (ROC) curve analysis. The serum levels of S100A12 at baseline were significantly elevated in patients in the ISR group compared with those in the non-ISR group (P<0.001). In the multivariate logistic regression analysis, after adjusting for conventional cardiovascular risk factors, laboratory parameters and medication after the procedure, the S100A12 level was revealed to be independently associated with ISR. When a cut-off for serum S100A12 levels of 34.75 ng/ml was used, the ROC curve was able to predict ISR with 72.8% sensitivity and 79.1% specificity, and the area under the ROC curve was 0.796 (95% CI: 0.757 to 0.834, P<0.001). Furthermore, addition of S100A12 to established risk factors significantly improved the predictive power of reference models for ISR. S100A12 may serve as an independent marker to predict ISR in patients undergoing coronary DES implantation.
Collapse
Affiliation(s)
- Hengyi Liang
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yuqi Cui
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, P.R. China.,Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65211, USA
| | - Haoran Bu
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hang Liu
- Deparment of Cardiology, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450014, P.R. China
| | - Pengcheng Yan
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Lianqun Cui
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Liming Chen
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
18
|
Iseri K, Dai L, Chen Z, Qureshi AR, Brismar TB, Stenvinkel P, Lindholm B. Bone mineral density and mortality in end-stage renal disease patients. Clin Kidney J 2020; 13:307-321. [PMID: 32699616 PMCID: PMC7367137 DOI: 10.1093/ckj/sfaa089] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis characterized by low bone mineral density (BMD) as assessed by dual-energy X-ray absorptiometry (DXA) is common among end-stage renal disease (ESRD) patients and associates with high fracture incidence and high all-cause mortality. This is because chronic kidney disease-mineral bone disorders (CKD-MBDs) promote not only bone disease (osteoporosis and renal dystrophy) but also vascular calcification and cardiovascular disease. The disturbed bone metabolism in ESRD leads to 'loss of cortical bone' with increased cortical porosity and thinning of cortical bone rather than to loss of trabecular bone. Low BMD, especially at cortical-rich bone sites, is closely linked to CKD-MBD, vascular calcification and poor cardiovascular outcomes. These effects appear to be largely mediated by shared mechanistic pathways via the 'bone-vascular axis' through which impaired bone status associates with changes in the vascular wall. Thus, bone is more than just the scaffolding that holds the body together and protects organs from external forces but is-in addition to its physical supportive function-also an active endocrine organ that interacts with the vasculature by paracrine and endocrine factors through pathways including Wnt signalling, osteoprotegerin (OPG)/receptor activator of nuclear factor-κB (RANK)/RANK ligand system and the Galectin-3/receptor of advanced glycation end products axis. The insight that osteogenesis and vascular calcification share many similarities-and the knowledge that vascular calcification is a cell-mediated active rather than a passive mineralization process-suggest that low BMD and vascular calcification ('vascular ossification') to a large extent represent two sides of the same coin. Here, we briefly review changes of BMD in ESRD as observed using different DXA methods (central and whole-body DXA) at different bone sites for BMD measurements, and summarize recent knowledge regarding the relationships between 'low BMD' and 'fracture incidence, vascular calcification and increased mortality' in ESRD patients, as well as potential 'molecular mechanisms' underlying these associations.
Collapse
Affiliation(s)
- Ken Iseri
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Division of Nephrology, Showa University School of Medicine, Tokyo, Japan
| | - Lu Dai
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Zhimin Chen
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Abdul Rashid Qureshi
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Torkel B Brismar
- Department of Clinical Science, Intervention and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Karolinska University Hospital, Huddinge, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Lindholm
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
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: 47] [Impact Index Per Article: 11.8] [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
|
20
|
Weber JR, Martin B, Kassis N, Shah K, Kovarnik T, Mattix-Kramer H, Lopez JJ. An optical coherence tomography comparison of coronary arterial plaque calcification in patients with end-stage renal disease and diabetes mellitus. Diab Vasc Dis Res 2020; 17:1479164120958425. [PMID: 32981349 PMCID: PMC7919205 DOI: 10.1177/1479164120958425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Coronary arterial plaques in patients with end-stage renal disease (ESRD) are assumed to have increased calcification due to underlying renal disease or initiation of dialysis. This relationship may be confounded by comorbid type 2 diabetes mellitus (DM). METHODS From a single-center OCT registry, 60 patients were analyzed. Twenty patients with ESRD and diabetes (ESRD-DM) were compared to 2 groups of non-ESRD patients: 20 with and 20 without diabetes. In each patient, one 20 mm segment within the culprit vessel was analyzed. RESULTS ESRD-DM patients exhibited similar calcium burden, arc, and area compared to patients with diabetes alone. When compared to patients without diabetes, patients with diabetes exhibited a greater summed area of calcium (DM: Median 9.0, IQR [5.3-28] mm2 vs Non-DM: 3.5 [0.1-14] mm2, p = 0.04) and larger calcium deposits by arc (DM: Mean 45 ± SE 6.2° vs Non-DM: 21 ± 6.2°, p = 0.01) and area (DM: 0.58 ± 0.10 mm2 vs Non-DM: 0.26 ± 0.10 mm2, p = 0.03). Calcification deposits in ESRD-DM patients (0.14 ± 0.02 mm) and patients with diabetes (0.14 ± 0.02 mm) were more superficially located relative to patients without diabetes (0.21 ± 0.02 mm), p = 0.01 for both. CONCLUSIONS Coronary calcification in DM and ESRD-DM groups exhibited similar burden, deposit size, and depth within the arterial wall. The increase in coronary calcification and cardiovascular disease events seen in ESRD-DM patients may not be secondary to ESRD and dialysis, but instead due to a combination of declining renal function and diabetes.
Collapse
Affiliation(s)
- Joseph R Weber
- Department of Medicine, Division of Cardiology, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Brendan Martin
- Department of Medicine, Division of Cardiology, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Nicholas Kassis
- Department of Medicine, Division of Cardiology, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Kunal Shah
- Department of Medicine, Division of Cardiology, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Tomas Kovarnik
- 2nd Department of Internal Medicine and Department of Cardiovascular Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Holly Mattix-Kramer
- Department of Health Sciences and Department of Medicine, Division of Nephrology and Hypertension, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - John J Lopez
- Department of Medicine, Division of Cardiology, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
21
|
Belmokhtar K, Ortillon J, Jaisson S, Massy ZA, Boulagnon Rombi C, Doué M, Maurice P, Fritz G, Gillery P, Schmidt AM, Rieu P, Touré F. Receptor for advanced glycation end products: a key molecule in the genesis of chronic kidney disease vascular calcification and a potential modulator of sodium phosphate co-transporter PIT-1 expression. Nephrol Dial Transplant 2020; 34:2018-2030. [PMID: 30778553 DOI: 10.1093/ndt/gfz012] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/05/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is associated with increased cardiovascular mortality, frequent vascular calcification (VC) and accumulation of uraemic toxins. Advanced glycation end products and S100 proteins interact with the receptor for advanced glycation end products (RAGE). In the present work, we aimed to investigate the role(s) of RAGE in the CKD-VC process. METHODS Apoe-/- or Apoe-/-Ager (RAGE)-/- male mice were assigned to CKD or sham-operated groups. A high-phosphate diet was given to a subgroup of Apoe-/-and Apoe-/-Ager-/- CKD mice. Primary cultures of Ager+/+ and Ager-/- vascular smooth muscle cells (VSMCs) were established and stimulated with either vehicle, inorganic phosphate (Pi) or RAGE ligands (S100A12; 20 µM). RESULTS After 12 weeks of CKD we observed a significant increase in RAGE ligand (AGE and S100 proteins) concentrations in the serum of CKD Apoe-/- mice. Ager messenger RNA (mRNA) levels were 4-fold higher in CKD vessels of Apoe-/- mice. CKD Apoe-/- but not CKD Apoe-/- or Ager-/- mice displayed a marked increase in the VC surface area. Similar trends were found in the high-phosphate diet condition. mRNA levels of Runx2 significantly increased in the Apoe-/- CKD group. In vitro, stimulation of Ager+/+VSMCs with Pi or S100A12 induced mineralization and osteoblast transformation, and this was inhibited by phosphonoformic acid (Pi co-transporters inhibitor) and Ager deletion. In vivo and in vitro RAGE was necessary for regulation of the expression of Pit-1, at least in part through production of reactive oxygen species. CONCLUSION RAGE, through the modulation of Pit-1 expression, is a key molecule in the genesis of VC.
Collapse
Affiliation(s)
- Karim Belmokhtar
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,Laboratoire de Néphrologie, Univesrity of Reims, Faculté de Médecine, Reims, France
| | - Jeremy Ortillon
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,Laboratoire de Néphrologie, Univesrity of Reims, Faculté de Médecine, Reims, France
| | - Stéphane Jaisson
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,University Hospital of Reims, Maison Blanche Hospital, Laboratory of Pediatric Biology and Research, Reims, France
| | - Ziad A Massy
- Division of Nephrology, Ambroise Paré Hospital, APHP, Versailles Saint-Quentin-en-Yvelines University (Paris-Ile-de-France-Ouest University), UVSQ, Boulogne Billancourt/Paris, France.,Inserm U1018, Team5, CESP, Paris Saclay Unioversityand Versailles Saint-Quentin-en-Yvelines University (Paris-Ile-de-France-Ouest University, UVSQ), Villejuif, France
| | - Camille Boulagnon Rombi
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,CHU Reims, Division of Anatomopathology, Reims, France
| | - Manon Doué
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Pascal Maurice
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France
| | - Günter Fritz
- Institute of Neuropathology, University of Freiburg, Germany
| | - Philippe Gillery
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,University Hospital of Reims, Maison Blanche Hospital, Laboratory of Pediatric Biology and Research, Reims, France
| | | | - Philippe Rieu
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,Laboratoire de Néphrologie, Univesrity of Reims, Faculté de Médecine, Reims, France.,CHU Reims, Division of Nephrology, Reims, France
| | - Fatouma Touré
- Université de Reims Champagne-Ardenne, CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Reims, France.,Laboratoire de Néphrologie, Univesrity of Reims, Faculté de Médecine, Reims, France.,CHU Reims, Division of Nephrology, Reims, France
| |
Collapse
|
22
|
S100 proteins in atherosclerosis. Clin Chim Acta 2020; 502:293-304. [DOI: 10.1016/j.cca.2019.11.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023]
|
23
|
Mukai H, Svedberg O, Lindholm B, Dai L, Heimbürger O, Barany P, Anderstam B, Stenvinkel P, Qureshi AR. Skin autofluorescence, arterial stiffness and Framingham risk score as predictors of clinical outcome in chronic kidney disease patients: a cohort study. Nephrol Dial Transplant 2019; 34:442-448. [PMID: 29378035 DOI: 10.1093/ndt/gfx371] [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] [Received: 09/14/2017] [Accepted: 12/18/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The risk of cardiovascular disease (CVD) is predicted by Framingham's CVD risk scores (FRS) but the high CVD-related mortality in patients with chronic kidney disease (CKD) is only partially explained by traditional CVD risk markers. Therefore, there is a need to explore whether other CVD risk markers may improve risk prediction. Although arterial stiffness measured by augmentation index (AIx) and tissue content of advanced glycation end-products (AGEs) measured by skin autofluorescence (SAF) are two biomarkers that associate with CVD and mortality in CKD, it is not known how they compare with FRS. We evaluated associations between SAF, AIx and FRS, and their associations with CVD and mortality in CKD patients. METHODS SAF (AGE Reader) and AIx (SphygmoCor; adjusted for 75 heart beats per minute) were measured in 261 clinically stable and extensively phenotyped patients with CKD Stage 5 (median age 56 years, 66% male, 20% diabetes; 130 non-dialysed, 93 patients on peritoneal dialysis and 38 patients on haemodialysis). Multivariate receiver operator characteristics (ROC) curve analysis and multivariate Cox models followed by C-statistics were used to evaluate CVD-related and all-cause mortality risk associated with SAF, AIx and FRS during follow-up for median 25 months with 46 deaths. RESULTS In multivariate regression analysis, SAF associated with FRS, haemoglobin, fat body mass index and CVD, and inversely with per cent handgrip strength (HGS). AIx associated with FRS, and inversely with per cent HGS. Associations of SAF and AIx with high-sensitivity C-reactive protein (hsCRP), serum albumin, statin therapy and renal replacement therapy were not statistically significant. In ROC analysis, area under the curve (AUC) for CVD mortality ranged from AUC = 0.72 (AIx and FRS, respectively) to AUC = 0.78 (FRS + AIx), and for all-cause mortality from AUC = 0.70 (AIx) to AUC = 0.79 (FRS + AIx). In multivariate Cox analysis, after adjusting for 1-standard deviation (1-SD) of FRS, 1-SD increase of SAF associated with all-cause mortality and 1-SD increase of AIx associated with CVD mortality and all-cause mortality. After further adjustments for hsCRP, albumin and presence of CVD, AIx (but not SAF) remained independently associated with CVD mortality, hazard ratio (HR) 2.14 [95% confidence interval (95% CI) 1.18-3.89] and all-cause mortality, HR 1.74 (95% CI 1.16-2.60). CONCLUSIONS In patients with CKD Stage 5, SAF and aortic stiffness associated with mortality, independently of FRS. After adjusting for additional confounders including inflammation, aortic stiffness remained as an independent predictor of outcome. Since the contribution of SAF and aortic stiffness compared with FRS in ROC curve analysis was relatively modest, this underlines the importance of traditional CVD risk factors in CKD.
Collapse
Affiliation(s)
- Hideyuki Mukai
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Oskar Svedberg
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Lu Dai
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Olof Heimbürger
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Peter Barany
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Björn Anderstam
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| |
Collapse
|
24
|
Basatemur GL, Jørgensen HF, Clarke MCH, Bennett MR, Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nat Rev Cardiol 2019; 16:727-744. [PMID: 31243391 DOI: 10.1038/s41569-019-0227-9] [Citation(s) in RCA: 609] [Impact Index Per Article: 121.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2019] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are a major cell type present at all stages of an atherosclerotic plaque. According to the 'response to injury' and 'vulnerable plaque' hypotheses, contractile VSMCs recruited from the media undergo phenotypic conversion to proliferative synthetic cells that generate extracellular matrix to form the fibrous cap and hence stabilize plaques. However, lineage-tracing studies have highlighted flaws in the interpretation of former studies, revealing that these studies had underestimated both the content and functions of VSMCs in plaques and have thus challenged our view on the role of VSMCs in atherosclerosis. VSMCs are more plastic than previously recognized and can adopt alternative phenotypes, including phenotypes resembling foam cells, macrophages, mesenchymal stem cells and osteochondrogenic cells, which could contribute both positively and negatively to disease progression. In this Review, we present the evidence for VSMC plasticity and summarize the roles of VSMCs and VSMC-derived cells in atherosclerotic plaque development and progression. Correct attribution and spatiotemporal resolution of clinically beneficial and detrimental processes will underpin the success of any therapeutic intervention aimed at VSMCs and their derivatives.
Collapse
Affiliation(s)
- Gemma L Basatemur
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Helle F Jørgensen
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Murray C H Clarke
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.
- INSERM U970, Paris Cardiovascular Research Center, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
25
|
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: 629] [Impact Index Per Article: 125.8] [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
|
26
|
Grauen Larsen H, Yndigegn T, Marinkovic G, Grufman H, Mares R, Nilsson J, Goncalves I, Schiopu A. The soluble receptor for advanced glycation end-products (sRAGE) has a dual phase-dependent association with residual cardiovascular risk after an acute coronary event. Atherosclerosis 2019; 287:16-23. [PMID: 31181415 DOI: 10.1016/j.atherosclerosis.2019.05.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/18/2019] [Accepted: 05/23/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND AIMS The pro-inflammatory alarmin S100A12 (EN-RAGE) and the soluble form of its receptor, the receptor for advanced glycation endproducts (sRAGE), have diverging roles in cardiovascular disease. In experimental studies, S100A12 promoted atherosclerosis while sRAGE treatment was anti-atherogenic and reduced myocardial infarction size by scavenging RAGE ligands. Here, we aimed to explore the links between S100A12, sRAGE, and long-term prognosis after an acute coronary syndrome (ACS). METHODS We measured S100A12 and sRAGE in 524 patients within 24 h after an ACS, and again 6 weeks later in a subgroup of 114 patients. This subgroup also completed a follow-up echocardiography after 1 year. The median follow-up time for recurrent major adverse cardiovascular events (MACE), defined as recurrent ACS or cardiovascular death, was 25.7 ± 12.6 months. RESULTS In Cox proportional hazard analyses, baseline S100A12 and sRAGE were positively associated with the risk of MACE, independently of traditional cardiovascular risk factors. The association between sRAGE and MACE remained significant after additional adjustment for troponin T, NT-proBNP and hsCRP [HR 95%CI for highest versus lowest tertile 3.2 (1.5-6.5), p = 0.002]. High sRAGE was also associated with deteriorating left ventricular function and an increased rate of heart failure hospitalization post-discharge. In contrast, patients with increasing sRAGE at 6 weeks compared to baseline had lower incidence of recurrent ACS. CONCLUSIONS Our data suggest that sRAGE has a dual, phase-dependent association with residual cardiovascular risk after ACS. These findings are important for the design and interpretation of future studies on sRAGE as biomarker and potential treatment in ACS patients.
Collapse
Affiliation(s)
- Helena Grauen Larsen
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden; Department of Cardiology, Skane University Hospital Malmö, Sweden
| | - Troels Yndigegn
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden
| | - Goran Marinkovic
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden
| | - Helena Grufman
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden
| | - Razvan Mares
- University of Medicine, Pharmacy, Sciences and Technology of Tirgu-Mures, 38 Gh. Marinescu Str., Tirgu-Mures, Romania
| | - Jan Nilsson
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden
| | - Isabel Goncalves
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden; Department of Cardiology, Skane University Hospital Malmö, Sweden
| | - Alexandru Schiopu
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden; Department of Cardiology, Skane University Hospital Malmö, Sweden; University of Medicine, Pharmacy, Sciences and Technology of Tirgu-Mures, 38 Gh. Marinescu Str., Tirgu-Mures, Romania.
| |
Collapse
|
27
|
Ngai D, Lino M, Bendeck MP. Cell-Matrix Interactions and Matricrine Signaling in the Pathogenesis of Vascular Calcification. Front Cardiovasc Med 2018; 5:174. [PMID: 30581820 PMCID: PMC6292870 DOI: 10.3389/fcvm.2018.00174] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification is a complex pathological process occurring in patients with atherosclerosis, type 2 diabetes, and chronic kidney disease. The extracellular matrix, via matricrine-receptor signaling plays important roles in the pathogenesis of calcification. Calcification is mediated by osteochondrocytic-like cells that arise from transdifferentiating vascular smooth muscle cells. Recent advances in our understanding of the plasticity of vascular smooth muscle cell and other cells of mesenchymal origin have furthered our understanding of how these cells transdifferentiate into osteochondrocytic-like cells in response to environmental cues. In the present review, we examine the role of the extracellular matrix in the regulation of cell behavior and differentiation in the context of vascular calcification. In pathological calcification, the extracellular matrix not only provides a scaffold for mineral deposition, but also acts as an active signaling entity. In recent years, extracellular matrix components have been shown to influence cellular signaling through matrix receptors such as the discoidin domain receptor family, integrins, and elastin receptors, all of which can modulate osteochondrocytic differentiation and calcification. Changes in extracellular matrix stiffness and composition are detected by these receptors which in turn modulate downstream signaling pathways and cytoskeletal dynamics, which are critical to osteogenic differentiation. This review will focus on recent literature that highlights the role of cell-matrix interactions and how they influence cellular behavior, and osteochondrocytic transdifferentiation in the pathogenesis of cardiovascular calcification.
Collapse
Affiliation(s)
- David Ngai
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Marsel Lino
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Chellan B, Sutton NR, Hofmann Bowman MA. S100/RAGE-Mediated Inflammation and Modified Cholesterol Lipoproteins as Mediators of Osteoblastic Differentiation of Vascular Smooth Muscle Cells. Front Cardiovasc Med 2018; 5:163. [PMID: 30467547 PMCID: PMC6235906 DOI: 10.3389/fcvm.2018.00163] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/19/2018] [Indexed: 01/01/2023] Open
Abstract
Arterial calcification is a feature of atherosclerosis and shares many risk factors including diabetes, dyslipidemia, chronic kidney disease, hypertension, and age. Although there is overlap in risk factors, anti-atherosclerotic therapies, including statins, fail to reduce arterial, and aortic valve calcifications. This suggests that low density lipoprotein (LDL) may not be the main driver for aortic valve disease and arterial calcification. This review focuses on modified LDLs and their role in mediating foam cell formation in smooth muscle cells (SMCs), with special emphasis on enzyme modified non-oxidized LDL (ELDL). In vivo, ELDL represents one of the many forms of modified LDLs present in the atherosclerotic vessel. Phenotypic changes of macrophages and SMCs brought about by the uptake of modified LDLs overlap significantly in an atherosclerotic milieu, making it practically impossible to differentiate between the effects from oxidized LDL, ELDL, and other LDL modification. By studying in vitro-generated modifications of LDL, we were able to demonstrate marked differences in the transcriptome of human coronary artery SMCs (HCASMCs) upon uptake of ELDL, OxLDL, and native LDL, indicating that specific modifications of LDL in atherosclerotic plaques may determine the biology and functional consequences in vasculature. Enzyme-modified non-oxidized LDL (ELDL) induces calcification of SMCs and this is associated with reduced mRNA levels for genes protective for calcification (ENPP1, MGP) and upregulation of osteoblastic genes. A second focus of this review is on the synergy between hyperlipidemia and accelerated calcification In vivo in a mouse models with transgenic expression of human S100A12. We summarize mechanisms of S100A12/RAGE mediated vascular inflammation promoting vascular and valve calcification in vivo.
Collapse
Affiliation(s)
- Bijoy Chellan
- Department of Medicine, University of Illinois, Chicago, IL, United States
| | - Nadia R Sutton
- Department of Medicine, University of Michigan, Ann Arbor, MI, United States
| | | |
Collapse
|
29
|
Nishida M, Saegusa J, Tanaka S, Morinobu A. S100A12 facilitates osteoclast differentiation from human monocytes. PLoS One 2018; 13:e0204140. [PMID: 30235276 PMCID: PMC6147475 DOI: 10.1371/journal.pone.0204140] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 09/03/2018] [Indexed: 12/29/2022] Open
Abstract
Osteoclasts play a critical role not only in bone homeostasis but also in inflammatory osteolysis, such as that occurring in inflammatory arthritis and systemic inflammation. In both inflammation conditions, inflammatory cytokines like Interleukin (IL)-1, IL-6 and tumor necrosis factor (TNF)-α induce RANKL expression in osteoblasts, but the roles of these cytokines in osteoclast activation remain unclear. S100A12, an S100 family member, is a low-molecular-weight calcium-binding protein. Although it has a pro-inflammatory role, its effects on osteoclast differentiation have been unclear. Here we examined the direct effects of S100A12 on human osteoclasts in vitro. S100A12 facilitated osteoclast formation in the presence of RANKL, as judged by the cells’ morphology and elevated expression of osteoclast-related molecules, including NFATc1, ACP5, CALCR, and ITGβ3. In addition, S100A12 administration markedly enhanced the osteoclasts’ bone resorption ability, consistent with their increased expression levels of CTSK and CA2. Blocking RAGE and TLR4 cancelled the effects of S100A12. Our results indicate that S100A12 is a potential therapeutic target for inflammatory osteolysis.
Collapse
Affiliation(s)
- Miwa Nishida
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
- The Center for Rheumatic Diseases, Shinko Hospital, Chuo-ku, Kobe, Japan
| | - Jun Saegusa
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
- Clinical Laboratory, Kobe University Hospital, Chuo-ku, Kobe, Japan
| | - Shino Tanaka
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
- * E-mail:
| |
Collapse
|
30
|
The Impact of Uremic Toxins on Vascular Smooth Muscle Cell Function. Toxins (Basel) 2018; 10:toxins10060218. [PMID: 29844272 PMCID: PMC6024314 DOI: 10.3390/toxins10060218] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/26/2018] [Accepted: 05/27/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with profound vascular remodeling, which accelerates the progression of cardiovascular disease. This remodeling is characterized by intimal hyperplasia, accelerated atherosclerosis, excessive vascular calcification, and vascular stiffness. Vascular smooth muscle cell (VSMC) dysfunction has a key role in the remodeling process. Under uremic conditions, VSMCs can switch from a contractile phenotype to a synthetic phenotype, and undergo abnormal proliferation, migration, senescence, apoptosis, and calcification. A growing body of data from experiments in vitro and animal models suggests that uremic toxins (such as inorganic phosphate, indoxyl sulfate and advanced-glycation end products) may directly impact the VSMCs’ physiological functions. Chronic, low-grade inflammation and oxidative stress—hallmarks of CKD—are also strong inducers of VSMC dysfunction. Here, we review current knowledge about the impact of uremic toxins on VSMC function in CKD, and the consequences for pathological vascular remodeling.
Collapse
|
31
|
Fuery MA, Liang L, Kaplan FS, Mohler ER. Vascular ossification: Pathology, mechanisms, and clinical implications. Bone 2018; 109:28-34. [PMID: 28688892 DOI: 10.1016/j.bone.2017.07.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/04/2017] [Accepted: 07/04/2017] [Indexed: 12/28/2022]
Abstract
In recent years, the mechanisms and clinical significance of vascular calcification have been increasingly investigated. For over a century, however, pathologists have recognized that vascular calcification is a form of heterotopic ossification. In this review, we aim to describe the pathology and molecular processes of vascular ossification, to characterize its clinical significance and treatment options, and to elucidate areas that require further investigation. The molecular mechanisms of vascular ossification involve the activation of regulators including bone morphogenic proteins and chondrogenic transcription factors and the loss of mineralization inhibitors like fetuin-A and pyrophosphate. Although few studies have examined the gross pathology of vascular ossification, the presence of these molecular regulators and evidence of microfractures and cartilage have been demonstrated on heart valves and atherosclerotic plaques. These changes are often triggered by common inflammatory and metabolic disorders like diabetes, hyperlipidemia, and chronic kidney disease. The increasing prevalence of these diseases warrants further research into the clinical significance of vascular ossification and future treatment options.
Collapse
Affiliation(s)
- Michael A Fuery
- Department of Medicine, Cardiovascular Division, Section of Vascular Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Lusha Liang
- Department of Medicine, Cardiovascular Division, Section of Vascular Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Frederick S Kaplan
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Emile R Mohler
- Department of Medicine, Cardiovascular Division, Section of Vascular Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
32
|
Pérez-Hernández N, Aptilon-Duque G, Blachman-Braun R, Vargas-Alarcón G, Rodríguez-Cortés AA, Azrad-Daniel S, Posadas-Sánchez R, Rodríguez-Pérez JM. Vascular Calcification: Current Genetics Underlying This Complex Phenomenon. Chin Med J (Engl) 2018; 130:1113-1121. [PMID: 28469108 PMCID: PMC5421183 DOI: 10.4103/0366-6999.204931] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE Vascular calcification is the consequence of the complex interaction between genetic, environmental, and vascular factors, which ultimately lead to the deposition of calcium in the tunica intima (atherosclerotic calcification) or tunica media (Mönckenberg's sclerosis). Vascular calcification is also closely related to other pathologies, such as diabetes mellitus, dyslipidemia, and chronic kidney disease. It has been concluded that the degree of vascular calcification may vary from person to person, even if the associated pathologies and environmental factors are the same. Therefore, this suggests an important genetic contribution to the development of vascular calcification. This review aimed to find the most recent evidence about vascular calcification pathophysiology regarding the genetic aspects and molecular pathways. DATA SOURCES We conducted an exhaustive search in Scopus, EBSCO, and PubMed with the keywords "genetics and vascular calcification", "molecular pathways, genetic and vascular calcification" and included the main articles from January 1995 up to August 2016. We focused on the most recent evidence about vascular calcification pathophysiology regarding the genetic aspects and molecular pathways. STUDY SELECTION The most valuable published original and review articles related to our objective were selected. RESULTS Vascular calcification is a multifactorial disease; thus, its pathophysiology cannot be explained by a single specific factor, rather than by the result of the association of several genetic variants, molecular pathway interactions, and environmental factors that promote its development. CONCLUSION Although several molecular aspects of this mechanism have been elucidated, there is still a need for a better understanding of the factors that predispose to this disease.
Collapse
Affiliation(s)
- Nonanzit Pérez-Hernández
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", México, Mexico City 14080, México
| | - Gad Aptilon-Duque
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", México, Mexico City 14080, México
| | - Ruben Blachman-Braun
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", México, Mexico City 14080, México
| | - Gilberto Vargas-Alarcón
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", México, Mexico City 14080, México
| | - Adrián Asael Rodríguez-Cortés
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", México, Mexico City 14080, México
| | - Shely Azrad-Daniel
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", México, Mexico City 14080, México
| | - Rosalinda Posadas-Sánchez
- Department of Endocrinology, National Institute of Cardiology "Ignacio Chávez", México, México City 14080, México
| | - José Manuel Rodríguez-Pérez
- Department of Molecular Biology, National Institute of Cardiology "Ignacio Chávez", México, Mexico City 14080, México
| |
Collapse
|
33
|
Panda DK, Bai X, Sabbagh Y, Zhang Y, Zaun HC, Karellis A, Koromilas AE, Lipman ML, Karaplis AC. Defective interplay between mTORC1 activity and endoplasmic reticulum stress-unfolded protein response in uremic vascular calcification. Am J Physiol Renal Physiol 2018; 314:F1046-F1061. [PMID: 29357413 DOI: 10.1152/ajprenal.00350.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vascular calcification increases the risk of cardiovascular disease and death in patients with chronic kidney disease (CKD). Increased activity of mammalian target of rapamycin complex 1 (mTORC1) and endoplasmic reticulum (ER) stress-unfolded protein response (UPR) are independently reported to partake in the pathogenesis of vascular calcification in CKD. However, the association between mTORC1 activity and ER stress-UPR remains unknown. We report here that components of the uremic state [activation of the receptor for advanced glycation end products (RAGE) and hyperphosphatemia] potentiate vascular smooth muscle cell (VSMC) calcification by inducing persistent and exaggerated activity of mTORC1. This gives rise to prolonged and excessive ER stress-UPR as well as attenuated levels of sestrin 1 ( Sesn1) and Sesn3 feeding back to inhibit mTORC1 activity. Activating transcription factor 4 arising from the UPR mediates cell death via expression of CCAAT/enhancer-binding protein (c/EBP) homologous protein (CHOP), impairs the generation of pyrophosphate, a potent inhibitor of mineralization, and potentiates VSMC transdifferentiation to the osteochondrocytic phenotype. Short-term treatment of CKD mice with rapamycin, an inhibitor of mTORC1, or tauroursodeoxycholic acid, a bile acid that restores ER homeostasis, normalized mTORC1 activity, molecular markers of UPR, and calcium content of aortas. Collectively, these data highlight that increased and/or protracted mTORC1 activity arising from the uremic state leads to dysregulated ER stress-UPR and VSMC calcification. Manipulation of the mTORC1-ER stress-UPR pathway opens up new therapeutic strategies for the prevention and treatment of vascular calcification in CKD.
Collapse
Affiliation(s)
- Dibyendu K Panda
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Xiuying Bai
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Yves Sabbagh
- Rare Disease, Sanofi Genzyme, Framingham, Massachusetts
| | - Yan Zhang
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Hans-Christian Zaun
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Angeliki Karellis
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Antonis E Koromilas
- Department of Oncology and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Mark L Lipman
- Division of Nephrology, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| | - Andrew C Karaplis
- Division of Endocrinology and Metabolism, Department of Medicine and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University , Montreal, Quebec , Canada
| |
Collapse
|
34
|
Iacobini C, Fantauzzi CB, Pugliese G, Menini S. Role of Galectin-3 in Bone Cell Differentiation, Bone Pathophysiology and Vascular Osteogenesis. Int J Mol Sci 2017; 18:ijms18112481. [PMID: 29160796 PMCID: PMC5713447 DOI: 10.3390/ijms18112481] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/17/2017] [Accepted: 11/19/2017] [Indexed: 01/15/2023] Open
Abstract
Galectin-3 is expressed in various tissues, including the bone, where it is considered a marker of chondrogenic and osteogenic cell lineages. Galectin-3 protein was found to be increased in the differentiated chondrocytes of the metaphyseal plate cartilage, where it favors chondrocyte survival and cartilage matrix mineralization. It was also shown to be highly expressed in differentiating osteoblasts and osteoclasts, in concomitance with expression of osteogenic markers and Runt-related transcription factor 2 and with the appearance of a mature phenotype. Galectin-3 is expressed also by osteocytes, though its function in these cells has not been fully elucidated. The effects of galectin-3 on bone cells were also investigated in galectin-3 null mice, further supporting its role in all stages of bone biology, from development to remodeling. Galectin-3 was also shown to act as a receptor for advanced glycation endproducts, which have been implicated in age-dependent and diabetes-associated bone fragility. Moreover, its regulatory role in inflammatory bone and joint disorders entitles galectin-3 as a possible therapeutic target. Finally, galectin-3 capacity to commit mesenchymal stem cells to the osteoblastic lineage and to favor transdifferentiation of vascular smooth muscle cells into an osteoblast-like phenotype open a new area of interest in bone and vascular pathologies.
Collapse
Affiliation(s)
- Carla Iacobini
- Department of Clinical and Molecular Medicine, La Sapienza University, 00185 Rome, Italy.
| | | | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, La Sapienza University, 00185 Rome, Italy.
| | - Stefano Menini
- Department of Clinical and Molecular Medicine, La Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
35
|
Farokhzadian J, Mangolian Shahrbabaki P, Bagheri V. S100A12-CD36 axis: A novel player in the pathogenesis of atherosclerosis? Cytokine 2017; 122:154104. [PMID: 28756107 DOI: 10.1016/j.cyto.2017.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/16/2017] [Accepted: 07/14/2017] [Indexed: 12/15/2022]
Abstract
S100A12 is a member of the S100 family of EF-hand calcium-binding proteins and have a variety of intracellular and extracellular activities. It exerts its proinflammatory effects by binding to the receptor for advanced glycation end products (RAGE) and Toll-like receptor 4 (TLR4). CD36 is a class B scavenger receptor that acts as a fatty acid transporter. Both S100A12 and CD36 are implicated in vascular inflammation and atherosclerosis. It has recently been demonstrated that S100A12 binds with high affinity to CD36. On the other hand, RAGE and TLR4 play a key role in the regulation of CD36 expression. These observations point to the fact that S100A12 is an interesting molecular target for the development of therapeutics. This Cytokine stimulus will focus on the possible mechanisms of S100A12-CD36 axis in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Jamileh Farokhzadian
- Nursing Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Community Health Nursing, School of Nursing and Midwifery, Kerman University of Medical Sciences, Kerman, Iran
| | - Parvin Mangolian Shahrbabaki
- Nursing Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Medical Surgical Nursing, School of Nursing and Midwifery, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Bagheri
- Immunology of Infectious Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
36
|
Perrucci GL, Zanobini M, Gripari P, Songia P, Alshaikh B, Tremoli E, Poggio P. Pathophysiology of Aortic Stenosis and Mitral Regurgitation. Compr Physiol 2017. [PMID: 28640443 DOI: 10.1002/cphy.c160020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The global impact of the spectrum of valve diseases is a crucial, fast-growing, and underrecognized health problem. The most prevalent valve diseases, requiring surgical intervention, are represented by calcific and degenerative processes occurring in heart valves, in particular, aortic and mitral valve. Due to the increasing elderly population, these pathologies will gain weight in the global health burden. The two most common valve diseases are aortic valve stenosis (AVS) and mitral valve regurgitation (MR). AVS is the most commonly encountered valve disease nowadays and affects almost 5% of elderly population. In particular, AVS poses a great challenge due to the multiple comorbidities and frailty of this patient subset. MR is also a common valve pathology and has an estimated prevalence of 3% in the general population, affecting more than 176 million people worldwide. This review will focus on pathophysiological changes in both these valve diseases, starting from the description of the anatomical aspects of normal valve, highlighting all the main cellular and molecular features involved in the pathological progression and cardiac consequences. This review also evaluates the main approaches in clinical management of these valve diseases, taking into account of the main published clinical guidelines. © 2017 American Physiological Society. Compr Physiol 7:799-818, 2017.
Collapse
Affiliation(s)
- Gianluca L Perrucci
- Centro Cardiologico Monzino, IRCCS, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | | | - Paola Songia
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | | - Paolo Poggio
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| |
Collapse
|
37
|
Beloqui O, Moreno MU, San José G, Pejenaute Á, Cortés A, Landecho MF, Díez J, Fortuño A, Zalba G. Increased phagocytic NADPH oxidase activity associates with coronary artery calcification in asymptomatic men. Free Radic Res 2017; 51:389-396. [PMID: 28427294 DOI: 10.1080/10715762.2017.1321745] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Vascular calcification is a common feature in atherosclerosis and associates with cardiovascular events. Oxidative stress may be involved in the pathogenesis of vascular calcification. Previous studies have shown that the phagocytic NADPH oxidase is associated with atherosclerosis. The objective of the present study was to investigate the association between phagocytic NADPH oxidase-mediated superoxide production and coronary artery calcium (CAC). NADPH oxidase-mediated superoxide production was determined by chemiluminescence and CAC by computed tomography in 159 asymptomatic men free of overt clinical atherosclerosis. Multivariate linear regression analyses were used to assess the relationship between CAC and NADPH oxidase-mediated superoxide production. Compared with individuals in the lowest score of CAC (= 0 Agatston units), those in the upper score (>400 Agatston units) showed higher superoxide production (p < 0.05). In correlation analysis, superoxide production positively (p < 0.01) correlated with CAC, which in multivariate analysis remained significant after adjusting for age, HDL-cholesterol, triglycerides, body mass index, smoking, arterial hypertension and diabetes mellitus. In conclusion, in a population of men without clinically overt atherosclerotic disease, increased NADPH oxidase-mediated superoxide production associated with enhanced CAC. Albeit descriptive, these findings suggest a potential involvement of phagocytic NADPH oxidase-mediated oxidative stress in CAC.
Collapse
Affiliation(s)
- Oscar Beloqui
- a Department of Internal Medicine , Clínica Universidad de Navarra , Pamplona , Spain.,b IdiSNA, Navarra Institute for Health Research , Pamplona , Spain
| | - María U Moreno
- b IdiSNA, Navarra Institute for Health Research , Pamplona , Spain.,c Division of Cardiovascular Pathophysiology , Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra , Pamplona , Spain
| | - Gorka San José
- b IdiSNA, Navarra Institute for Health Research , Pamplona , Spain.,c Division of Cardiovascular Pathophysiology , Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra , Pamplona , Spain
| | - Álvaro Pejenaute
- d Department of Biochemistry and Genetics , University of Navarra , Pamplona , Spain
| | - Adriana Cortés
- d Department of Biochemistry and Genetics , University of Navarra , Pamplona , Spain
| | - Manuel F Landecho
- a Department of Internal Medicine , Clínica Universidad de Navarra , Pamplona , Spain.,b IdiSNA, Navarra Institute for Health Research , Pamplona , Spain
| | - Javier Díez
- b IdiSNA, Navarra Institute for Health Research , Pamplona , Spain.,c Division of Cardiovascular Pathophysiology , Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra , Pamplona , Spain.,e Deparment of Cardiology and Cardiac Surgery , Clínica Universidad de Navarra , Pamplona , Spain
| | - Ana Fortuño
- b IdiSNA, Navarra Institute for Health Research , Pamplona , Spain.,c Division of Cardiovascular Pathophysiology , Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra , Pamplona , Spain
| | - Guillermo Zalba
- b IdiSNA, Navarra Institute for Health Research , Pamplona , Spain.,c Division of Cardiovascular Pathophysiology , Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra , Pamplona , Spain.,d Department of Biochemistry and Genetics , University of Navarra , Pamplona , Spain
| |
Collapse
|
38
|
Buyukterzi Z, Can U, Alpaydin S, Guzelant A, Karaarslan S, Kocyigit D, Gurses KM. Enhanced S100A9 and S100A12 expression in acute coronary syndrome. Biomark Med 2017; 11:229-237. [PMID: 28157385 DOI: 10.2217/bmm-2016-0253] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
AIMS In this study, we aimed to investigate whether serum S100A8, S100A9 and S100A12 levels were markers of acute coronary syndrome (ACS). MATERIALS & METHODS Patients who underwent coronary angiography and/or percutaneous coronary interventions between June 2015-October 2015 were consecutively recruited in this study and categorized three groups each containing 30 patients (normal coronary arteries, stable coronary artery disease, and acute coronary syndrome). Baseline characteristics, including co- morbidities and medications, were recorded and serum S100A8, S100A9, S100A12, and C- reactive protein levels were measured besides routine laboratory tests. RESULTS A total of 90 patients (63.00 [56.00-73.00] years, 62.89% male) have been included. None of the groups differed from each other regarding baseline characteristics (p > 0.05). S100A9 levels were elevated in ACS when compared with the normal coronary arteries (p = 0.033) and S100A12 levels were found to be elevated in ACS when compared with both patients with normal coronary arteries and stable coronary artery disease (p = 0.001). S100A12 was identified as an independent associate of ACS (p = 0.002). CONCLUSION These results suggest that S100A12 may serve as a marker of coronary plaque instability, and may have a therapeutic implication in ACS treatment.
Collapse
Affiliation(s)
- Zafer Buyukterzi
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Ummugulsum Can
- Department of Biochemistry, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Sertac Alpaydin
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Asuman Guzelant
- Department of Microbiology & Infectious Diseases, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Sukru Karaarslan
- Department of Cardiology, Ufuk University Faculty of Medicine, 06520 Balgat Ankara, Turkey
| | - Duygu Kocyigit
- Department of Cardiology, Hacettepe University Faculty of Medicine, 06100 Sihhiye Ankara, Turkey
| | - Kadri Murat Gurses
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| |
Collapse
|
39
|
Stabley JN, Towler DA. Arterial Calcification in Diabetes Mellitus: Preclinical Models and Translational Implications. Arterioscler Thromb Vasc Biol 2017; 37:205-217. [PMID: 28062508 PMCID: PMC5480317 DOI: 10.1161/atvbaha.116.306258] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus increasingly afflicts our aging and dysmetabolic population. Type 2 diabetes mellitus and the antecedent metabolic syndrome represent the vast majority of the disease burden-increasingly prevalent in children and older adults. However, type 1 diabetes mellitus is also advancing in preadolescent children. As such, a crushing wave of cardiometabolic disease burden now faces our society. Arteriosclerotic calcification is increased in metabolic syndrome, type 2 diabetes mellitus, and type 1 diabetes mellitus-impairing conduit vessel compliance and function, thereby increasing the risk for dementia, stroke, heart attack, limb ischemia, renal insufficiency, and lower extremity amputation. Preclinical models of these dysmetabolic settings have provided insights into the pathobiology of arterial calcification. Osteochondrogenic morphogens in the BMP-Wnt signaling relay and transcriptional regulatory programs driven by Msx and Runx gene families are entrained to innate immune responses-responses activated by the dysmetabolic state-to direct arterial matrix deposition and mineralization. Recent studies implicate the endothelial-mesenchymal transition in contributing to the phenotypic drift of mineralizing vascular progenitors. In this brief overview, we discuss preclinical disease models that provide mechanistic insights-and point to challenges and opportunities to translate these insights into new therapeutic strategies for our patients afflicted with diabetes mellitus and its arteriosclerotic complications.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Arteries/metabolism
- Arteries/pathology
- Atherosclerosis/etiology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Angiopathies/etiology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diet, High-Fat
- Disease Models, Animal
- Female
- Genetic Predisposition to Disease
- Humans
- Hyperlipidemias/complications
- Hyperlipidemias/genetics
- Male
- Phenotype
- Plaque, Atherosclerotic
- Rats
- Signal Transduction
- Translational Research, Biomedical
- Vascular Calcification/etiology
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- John N Stabley
- From the Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Dwight A Towler
- From the Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
40
|
Yahagi K, Kolodgie FD, Lutter C, Mori H, Romero ME, Finn AV, Virmani R. Pathology of Human Coronary and Carotid Artery Atherosclerosis and Vascular Calcification in Diabetes Mellitus. Arterioscler Thromb Vasc Biol 2016; 37:191-204. [PMID: 27908890 DOI: 10.1161/atvbaha.116.306256] [Citation(s) in RCA: 344] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 11/21/2016] [Indexed: 12/25/2022]
Abstract
The continuing increase in the prevalence of diabetes mellitus in the general population is predicted to result in a higher incidence of cardiovascular disease. Although the mechanisms of diabetes mellitus-associated progression of atherosclerosis are not fully understood, at clinical and pathological levels, there is an appreciation of increased disease burden and higher levels of arterial calcification in these subjects. Plaques within the coronary arteries of patients with diabetes mellitus generally exhibit larger necrotic cores and significantly greater inflammation consisting mainly of macrophages and T lymphocytes relative to patients without diabetes mellitus. Moreover, there is a higher incidence of healed plaque ruptures and positive remodeling in hearts from subjects with type 1 diabetes mellitus and type 2 diabetes mellitus, suggesting a more active atherogenic process. Lesion calcification in the coronary, carotid, and other arterial beds is also more extensive. Although the role of coronary artery calcification in identifying cardiovascular disease and predicting its outcome is undeniable, our understanding of how key hormonal and physiological alterations associated with diabetes mellitus such as insulin resistance and hyperglycemia influence the process of vascular calcification continues to grow. Important drivers of atherosclerotic calcification in diabetes mellitus include oxidative stress, endothelial dysfunction, alterations in mineral metabolism, increased inflammatory cytokine production, and release of osteoprogenitor cells from the marrow into the circulation. Our review will focus on the pathophysiology of type 1 diabetes mellitus- and type 2 diabetes mellitus-associated vascular disease with particular focus on coronary and carotid atherosclerotic calcification.
Collapse
Affiliation(s)
- Kazuyuki Yahagi
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Frank D Kolodgie
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Christoph Lutter
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Hiroyoshi Mori
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Maria E Romero
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Aloke V Finn
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Renu Virmani
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.).
| |
Collapse
|
41
|
Wagenknecht LE, Divers J, Register TC, Russell GB, Bowden DW, Xu J, Langefeld CD, Lenchik L, Hruska KA, Carr JJ, Freedman BI. Bone Mineral Density and Progression of Subclinical Atherosclerosis in African-Americans With Type 2 Diabetes. J Clin Endocrinol Metab 2016; 101:4135-4141. [PMID: 27552541 PMCID: PMC5095232 DOI: 10.1210/jc.2016-1934] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
CONTEXT Relative to European Americans, calcified atherosclerotic plaque (CP) is less prevalent and severe in African-Americans (AAs). OBJECTIVE Predictors of progression of CP in the aorta, carotid, and coronary arteries were examined in AAs over a mean 5.3 ± 1.4-year interval. DESIGN This is the African American-Diabetes Heart Study. SETTING A type 2 diabetes (T2D)-affected cohort was included. PARTICIPANTS A total of 300 unrelated AAs with T2D; 50% female, mean age 55 ± 9 years, baseline hemoglobin A1c 8.1 ± 1.8% was included. MAIN OUTCOME MEASURES Glycemic control, renal parameters, vitamin D, and computed tomography-derived measures of adiposity, vascular CP, and volumetric bone mineral density (vBMD) in lumbar and thoracic vertebrae were obtained at baseline and follow-up. RESULTS CP increased in incidence and quantity/mass in all three vascular beds over the 5-year study (P < .0001). Lower baseline lumbar and thoracic vBMD were associated with progression of abdominal aorta CP (P < .008), but not progression of carotid or coronary artery CP. Lower baseline estimated glomerular filtration rate was associated with progression of carotid artery CP (P = .0004), and higher baseline pericardial adipose volume was associated with progression of coronary artery (P = .001) and aorta (P = .0006) CP independent of body mass index. There was a trend for an inverse relationship between change in thoracic vBMD and change in aortic CP (P = .05). CONCLUSIONS In this longitudinal study, lower baseline thoracic and lumbar vBMD and estimated glomerular filtration rate and higher pericardial adipose volumes were associated with increases in CP in AAs with T2D. Changes in these variables and baseline levels and/or changes in glycemic control, albuminuria, and vitamin D were not significantly associated with progression of CP.
Collapse
Affiliation(s)
- Lynne E Wagenknecht
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jasmin Divers
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Thomas C Register
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Gregory B Russell
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Donald W Bowden
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jianzhao Xu
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Carl D Langefeld
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Leon Lenchik
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Keith A Hruska
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - J Jeffrey Carr
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Barry I Freedman
- Division of Public Health Sciences (L.E.W., J.D., G.B.R., C.D.L.), Department of Pathology (T.C.R.), Department of Biochemistry (D.W.B., J.X.), and Department of Radiology (L.L.), Wake Forest School of Medicine, Winston-Salem, North Carolina; Division of Pediatric Nephrology (K.A.H.), Washington University School of Medicine, St. Louis, Missouri; Department of Radiology (J.J.C.), Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Internal Medicine (B.I.F.), Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
42
|
HMGB1, IL-1α, IL-33 and S100 proteins: dual-function alarmins. Cell Mol Immunol 2016; 14:43-64. [PMID: 27569562 PMCID: PMC5214941 DOI: 10.1038/cmi.2016.34] [Citation(s) in RCA: 324] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/08/2023] Open
Abstract
Our immune system is based on the close collaboration of the innate and adaptive immune systems for the rapid detection of any threats to the host. Recognition of pathogen-derived molecules is entrusted to specific germline-encoded signaling receptors. The same receptors have now also emerged as efficient detectors of misplaced or altered self-molecules that signal tissue damage and cell death following, for example, disruption of the blood supply and subsequent hypoxia. Many types of endogenous molecules have been shown to provoke such sterile inflammatory states when released from dying cells. However, a group of proteins referred to as alarmins have both intracellular and extracellular functions which have been the subject of intense research. Indeed, alarmins can either exert beneficial cell housekeeping functions, leading to tissue repair, or provoke deleterious uncontrolled inflammation. This group of proteins includes the high-mobility group box 1 protein (HMGB1), interleukin (IL)-1α, IL-33 and the Ca2+-binding S100 proteins. These dual-function proteins share conserved regulatory mechanisms, such as secretory routes, post-translational modifications and enzymatic processing, that govern their extracellular functions in time and space. Release of alarmins from mesenchymal cells is a highly relevant mechanism by which immune cells can be alerted of tissue damage, and alarmins play a key role in the development of acute or chronic inflammatory diseases and in cancer development.
Collapse
|
43
|
Chen Q, Bei JJ, Liu C, Feng SB, Zhao WB, Zhou Z, Yu ZP, Du XJ, Hu HY. HMGB1 Induces Secretion of Matrix Vesicles by Macrophages to Enhance Ectopic Mineralization. PLoS One 2016; 11:e0156686. [PMID: 27243975 PMCID: PMC4887028 DOI: 10.1371/journal.pone.0156686] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/18/2016] [Indexed: 12/14/2022] Open
Abstract
Numerous clinical conditions have been linked to ectopic mineralization (EM). This process of pathological biomineralization is complex and not fully elucidated, but thought to be started within matrix vesicles (MVs). We hypothesized that high mobility group box 1 (HMGB1), a cytokine associated with biomineralizing process under physiological and pathological conditions, induces EM via promoting MVs secretion from macrophages. In this study, we found that HMGB1 significantly promoted secretion of MVs from macrophages and subsequently led to mineral deposition in elevated Ca/Pi medium in vitro. Transmission electron microscopy of calcifying MVs showed formation of hydroxyapatite crystals in the vesicle interior. Subcutaneous injection into mice with MVs derived from HMGB1-treated cells showed a greater potential to initiate regional mineralization. Mechanistic experiments revealed that HMGB1 activated neutral sphingomyelinase2 (nSMase2) that involved the receptor for advanced glycation end products (RAGE) and p38 MAPK (upstream of nSMase2). Inhibition of nSMase2 with GW4869 or p38 MAPK with SB-239063 prevented MVs secretion and mineral deposition. Collectively, HMGB1 induces MVs secretion from macrophages at least in part, via the RAGE/p38 MAPK/nSMase2 signaling pathway. Our findings thus reveal a novel mechanism by which HMGB1 induces ectopic mineralization.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Department of Out-patient, Naval University of Engineering, Wuhan, China
| | - Jun-Jie Bei
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chuan Liu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Shi-Bin Feng
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wei-Bo Zhao
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhou Zhou
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Zheng-Ping Yu
- Department of Occupational Health, Faculty of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Xiao-Jun Du
- Experimental Cardiology, Baker IDI Heart and Diabetes Institute, and Central Clinical School, Monash University, Melbourne, Australia
| | - Hou-Yuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
44
|
Abstract
Vascular disease, such as atherosclerosis and diabetic vasculopathy, is frequently complicated by vascular calcification. Previously believed to be an end-stage process of unregulated mineral precipitation, it is now well established to be a multi-faceted disease influenced by the characteristics of its vascular location, the origins of calcifying cells and numerous regulatory pathways. It reflects the fundamental plasticity of the vasculature that is gradually being revealed by progress in vascular and stem cell biology. This review provides a brief overview of where we stand in our understanding of vascular calcification, facing the challenge of translating this knowledge into viable preventive and therapeutic strategies.
Collapse
|
45
|
Fuentes E, Palomo I, Rojas A. Cross-talk between platelet and tumor microenvironment: Role of multiligand/RAGE axis in platelet activation. Blood Rev 2016; 30:213-21. [PMID: 26723842 DOI: 10.1016/j.blre.2015.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 10/02/2015] [Accepted: 11/30/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule R09I2001, Talca, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule R09I2001, Talca, Chile
| | - Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile.
| |
Collapse
|
46
|
Circulating S100A12 Levels Are Associated with Progression of Abdominal Aortic Calcification in Hemodialysis Patients. PLoS One 2016; 11:e0150145. [PMID: 26914918 PMCID: PMC4768003 DOI: 10.1371/journal.pone.0150145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 02/09/2016] [Indexed: 11/19/2022] Open
Abstract
Vascular calcification is an important factor associated with mortality in dialysis patients. Recently, soluble receptor for advanced glycation end product (sRAGE) and extracellular RAGE binding protein S100A12 (EN-RAGE) have been reported to be involved in the process of vascular calcification. Therefore, we investigated whether sRAGE and S100A12 are useful indicators of progression of abdominal aortic calcification in hemodialysis (HD) patients. We analyzed annual changes in vascular calcification score (VCS) for up to 4 years, compared to clinical and biological parameters in 149 HD patients. VCS was assessed annually using plain X-ray images of the lateral lumbar spine. The progression group was defined as patients with an increase in VCS more than 1 point each year on average during the observation period. Time-averaged concentrations were also evaluated to examine the association between biological parameters and changes in VCS. The patients had a mean age of 58.59 ± 12.93 years; 53.7% were male, and 45% were diabetic. The VCS increased in 55 patients; the mean increase was 1.60 ± 2.91 points. In a stepwise multivariate logistic analysis, we found that higher levels of S100A12 were significantly associated with progression of VCS (odds ratio [OR], 2.622; 95% confidence interval [CI], 1.371–5.016; P = 0.004). The relationship between sRAGE and VCS was not statistically significant (OR, 0.644; 95% CI, 0.302–1.374; P = 0.255). Our findings suggest that serum levels of S100A12 are associated with progression of abdominal aortic calcification in HD patients, independent of sRAGE level.
Collapse
|
47
|
Tang Y, Xu Q, Peng H, Liu Z, Yang T, Yu Z, Cheng G, Li X, Zhang G, Shi R. The role of vascular peroxidase 1 in ox-LDL-induced vascular smooth muscle cell calcification. Atherosclerosis 2015; 243:357-63. [DOI: 10.1016/j.atherosclerosis.2015.08.047] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 08/19/2015] [Accepted: 08/31/2015] [Indexed: 12/18/2022]
|
48
|
Oesterle A, Bowman MAH. S100A12 and the S100/Calgranulins: Emerging Biomarkers for Atherosclerosis and Possibly Therapeutic Targets. Arterioscler Thromb Vasc Biol 2015; 35:2496-507. [PMID: 26515415 DOI: 10.1161/atvbaha.115.302072] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/14/2015] [Indexed: 11/16/2022]
Abstract
Atherosclerosis is mediated by local and systematic inflammation. The multiligand receptor for advanced glycation end products (RAGE) has been studied in animals and humans and is an important mediator of inflammation and atherosclerosis. This review focuses on S100/calgranulin proteins (S100A8, S100A9, and S100A12) and their receptor RAGE in mediating vascular inflammation. Mice lack the gene for S100A12, which in humans is located on chromosome 3 between S100A8 and S100A9. Transgenic mice with smooth muscle cell-targeted expression of S100A12 demonstrate increased coronary and aortic calcification, as well as increased plaque vulnerability. Serum S100A12 has recently been shown to predict future cardiovascular events in a longitudinal population study, underscoring a role for S100A12 as a potential biomarker for coronary artery disease. Genetic ablation of S100A9 or RAGE in atherosclerosis-susceptible apolipoprotein E null mice results in reduced atherosclerosis. Importantly, S100A12 and the RAGE axis can be modified pharmacologically. For example, soluble RAGE reduces murine atherosclerosis and vascular inflammation. Additionally, a class of compounds currently in phase III clinical trials for multiple sclerosis and rheumatologic conditions, the quinoline-3-carboxamides, reduce atherosclerotic plaque burden and complexity in transgenic S100A12 apolipoprotein E null mice, but have not been tested with regards to human atherosclerosis. The RAGE axis is an important mediator for inflammation-induced atherosclerosis, and S100A12 has emerged as biomarker for human atherosclerosis. Decreasing inflammation by inhibiting S100/calgranulin-mediated activation of RAGE attenuates murine atherosclerosis, and future studies in patients with coronary artery disease are warranted to confirm S100/RAGE as therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Adam Oesterle
- From the Department of Medicine, The University of Chicago, IL
| | | |
Collapse
|
49
|
Filsoof DM, Safford RE, Newby K, Rosenberg S, Kontras DG, Baker A, Odunukan OW, Fletcher G. Impact of exercise stress testing on diagnostic gene expression in patients with obstructive and nonobstructive coronary artery disease. Am J Cardiol 2015; 115:1346-50. [PMID: 25776454 DOI: 10.1016/j.amjcard.2015.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 11/30/2022]
Abstract
A blood-based gene expression test can diagnose obstructive coronary artery disease (CAD). The test is sensitive to inflammatory and immune processes associated with atherosclerosis. Acute exercise engages short-term inflammatory pathways, and exercise stress testing may affect results of gene expression testing during the same diagnostic workup. The objective of this study was to evaluate the effect of exercise on diagnostic gene expression testing. Ten patients with obstructive CAD (≥50% stenosis) and 10 with no/minimal CAD (≤20% stenosis) were identified by angiography. Blood samples for gene expression were obtained at baseline, peak exercise, 30 to 60 minutes after testing, and 24 to 36 hours after testing. Core-lab gene expression analysis yielded raw gene expression scores (GES) for each time point. Linear models were used to estimate changes in GES, adjusting for CAD status and other covariates. GES increased during peak exercise across both genders, with no significant differences as a function of CAD status. The overall adjusted mean GES increase at peak exercise was 0.29 (95% confidence interval 0.22 to 0.36; p <0.001). GES after exercise were not significantly different from baseline. The change in gene expression levels during peak exercise may reflect a transient inflammatory response to acute exercise that may be independent of patient gender or CAD status. In conclusion, CAD GES increase at peak exercise testing and rapidly return to baseline. Such may reflect a transient inflammatory response to acute exercise independent of gender or extent of CAD.
Collapse
Affiliation(s)
- David M Filsoof
- Division of Cardiovascular Diseases at Mayo Clinic, Jacksonville, Florida
| | - Robert E Safford
- Division of Cardiovascular Diseases at Mayo Clinic, Jacksonville, Florida
| | - Kristin Newby
- Division of Cardiology, Duke University, Durham, North Carolina
| | | | - Dana G Kontras
- Division of Cardiovascular Diseases at Mayo Clinic, Jacksonville, Florida
| | | | | | - Gerald Fletcher
- Division of Cardiovascular Diseases at Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
50
|
McCarty MF, DiNicolantonio JJ. The Molecular Biology and Pathophysiology of Vascular Calcification. Postgrad Med 2015; 126:54-64. [DOI: 10.3810/pgm.2014.03.2740] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|