1
|
Wang D, Li Q, Xie C. The role and mechanism of protein post‑translational modification in vascular calcification (Review). Exp Ther Med 2024; 28:419. [PMID: 39301258 PMCID: PMC11411399 DOI: 10.3892/etm.2024.12708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
Vascular calcification is closely associated with morbidity and mortality in patients with chronic kidney disease, atherosclerosis and diabetes. In the past few decades, vascular calcification has been studied extensively and the findings have shown that the mechanism of vascular calcification is not merely a consequence of a high-phosphorus and high-calcium environment but also an active process characterized by abnormal calcium phosphate deposition on blood vessel walls that involves various molecular mechanisms. Recent advances in bioinformatics approaches have led to increasing recognition that aberrant post-translational modifications (PTMs) play important roles in vascular calcification. This review presents the latest progress in clarifying the roles of PTMs, such as ubiquitination, acetylation, carbamylation and glycosylation, as well as signaling pathways, such as the Wnt/β-catenin pathway, in vascular calcification.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Medical Science, Yangzhou Polytechnic College, Yangzhou, Jiangsu 225100, P.R. China
| | - Qin Li
- Department of Medical Science, Yangzhou Polytechnic College, Yangzhou, Jiangsu 225100, P.R. China
| | - Caidie Xie
- Department of Nephrology, Nanjing Second Hospital, Nanjing Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210037, P.R. China
| |
Collapse
|
2
|
Tóth A, Balogh E, Jeney V. In Vitro Models of Cardiovascular Calcification. Biomedicines 2024; 12:2155. [PMID: 39335668 PMCID: PMC11429067 DOI: 10.3390/biomedicines12092155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/15/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular calcification, characterized by hydroxyapatite deposition in the arterial wall and heart valves, is associated with high cardiovascular morbidity and mortality. Cardiovascular calcification is a hallmark of aging but is frequently seen in association with chronic diseases, such as chronic kidney disease (CKD), diabetes, dyslipidemia, and hypertension in the younger population as well. Currently, there is no therapeutic approach to prevent or cure cardiovascular calcification. The pathophysiology of cardiovascular calcification is highly complex and involves osteogenic differentiation of various cell types of the cardiovascular system, such as vascular smooth muscle cells and valve interstitial cells. In vitro cellular and ex vivo tissue culture models are simple and useful tools in cardiovascular calcification research. These models contributed largely to the discoveries of the numerous calcification inducers, inhibitors, and molecular mechanisms. In this review, we provide an overview of the in vitro cell culture and the ex vivo tissue culture models applied in the research of cardiovascular calcification.
Collapse
Affiliation(s)
- Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Enikő Balogh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
3
|
Gheorghe SR, Crăciun AM, Ilyés T, Tisa IB, Sur L, Lupan I, Samasca G, Silaghi CN. Converging Mechanisms of Vascular and Cartilaginous Calcification. BIOLOGY 2024; 13:565. [PMID: 39194503 DOI: 10.3390/biology13080565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Physiological calcification occurs in bones and epiphyseal cartilage as they grow, whereas ectopic calcification occurs in blood vessels, cartilage, and soft tissues. Although it was formerly thought to be a passive and degenerative process associated with aging, ectopic calcification has been identified as an active cell-mediated process resembling osteogenesis, and an increasing number of studies have provided evidence for this paradigm shift. A significant association between vascular calcification and cardiovascular risk has been demonstrated by various studies, which have shown that arterial calcification has predictive value for future coronary events. With respect to cartilaginous calcification, calcium phosphate or hydroxyapatite crystals can form asymptomatic deposits in joints or periarticular tissues, contributing to the pathophysiology of osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, tendinitis, and bursitis. The risk factors and sequence of events that initiate ectopic calcification, as well as the mechanisms that prevent the development of this pathology, are still topics of debate. Consequently, in this review, we focus on the nexus of the mechanisms underlying vascular and cartilaginous calcifications, trying to circumscribe the similarities and disparities between them to provide more clarity in this regard.
Collapse
Affiliation(s)
- Simona R Gheorghe
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandra M Crăciun
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Tamás Ilyés
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Badiu Tisa
- Department of Pediatrics III, Iuliu Hatieganu University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania
| | - Lucia Sur
- Department of Pediatrics I, Iuliu Hatieganu University of Medicine and Pharmacy, 400370 Cluj-Napoca, Romania
| | - Iulia Lupan
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Ciprian N Silaghi
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
4
|
Wu S, Luo X, Chen Y, Wang Z, Liu X, Sun N, Zhao J, Luo W, Zhang J, Tong X, Huang L, Liu C, Qin Z. Sodium-glucose cotransporter 2 inhibitors attenuate vascular calcification by suppressing endoplasmic reticulum protein thioredoxin domain containing 5 dependent osteogenic reprogramming. Redox Biol 2024; 73:103183. [PMID: 38759418 PMCID: PMC11127605 DOI: 10.1016/j.redox.2024.103183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
AIMS Vascular calcification is strongly linked to the development of major adverse cardiovascular events, but effective treatments are lacking. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are an emerging category of oral hypoglycemic drugs that have displayed marked effects on metabolic and cardiovascular diseases, including recently reported vascular medial calcification. However, the roles and underlying mechanisms of SGLT2 inhibitors in vascular calcification have not been fully elucidated. Thus, we aimed to further determine whether SGLT2 inhibitors protect against vascular calcification and to investigate the mechanisms involved. METHODS AND RESULTS A computed tomography angiography investigation of coronary arteries from 1554 patients with type 2 diabetes revealed that SGLT2 inhibitor use was correlated with a lower Agatston calcification score. In the vitamin D3 overdose, 5/6 nephrectomy chronic kidney disease-induced medial calcification and Western diet-induced atherosclerotic intimal calcification models, dapagliflozin (DAPA) substantially alleviated vascular calcification in the aorta. Furthermore, we showed that DAPA reduced vascular calcification via Runx2-dependent osteogenic transdifferentiation in vascular smooth muscle cells (VSMCs). Transcriptome profiling revealed that thioredoxin domain containing 5 (TXNDC5) was involved in the attenuation of vascular calcification by DAPA. Rescue experiments showed that DAPA-induced TXNDC5 downregulation in VSMCs blocked the protective effect on vascular calcification. Furthermore, TXNDC5 downregulation disrupted protein folding-dependent Runx2 stability and promoted subsequent proteasomal degradation. Moreover, DAPA downregulated TXNDC5 expression via amelioration of oxidative stress and ATF6-dependent endoplasmic reticulum stress. Consistently, the class effects of SGLT2 inhibitors on vascular calcification were validated with empagliflozin in intimal and medial calcification models. CONCLUSIONS SGLT2 inhibitors ameliorate vascular calcification through blocking endoplasmic reticulum stress-dependent TXNDC5 upregulation and promoting subsequent Runx2 proteasomal degradation, suggesting that SGLT2 inhibitors are potentially beneficial for vascular calcification treatment and prevention.
Collapse
MESH Headings
- Vascular Calcification/metabolism
- Vascular Calcification/drug therapy
- Vascular Calcification/pathology
- Vascular Calcification/etiology
- Sodium-Glucose Transporter 2 Inhibitors/pharmacology
- Animals
- Humans
- Osteogenesis/drug effects
- Mice
- Glucosides/pharmacology
- Male
- Thioredoxins/metabolism
- Thioredoxins/genetics
- Benzhydryl Compounds/pharmacology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Endoplasmic Reticulum/metabolism
- Endoplasmic Reticulum/drug effects
- Rats
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Disease Models, Animal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Endoplasmic Reticulum Stress/drug effects
- Female
Collapse
Affiliation(s)
- Shaofa Wu
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China; Department of Nephrology, Youyang Hospital, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 409800, China
| | - Xiaolin Luo
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Yang Chen
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Zelan Wang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xi Liu
- Department of Nephrology, Youyang Hospital, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 409800, China
| | - Ning Sun
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Junyong Zhao
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Wenjian Luo
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jiawen Zhang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xiaoyong Tong
- Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China
| | - Lan Huang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| | - Chuan Liu
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| | - Zhexue Qin
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| |
Collapse
|
5
|
Ghosh S, Ghzaiel I, Vejux A, Meaney S, Nag S, Lizard G, Tripathi G, Naez F, Paul S. Impact of Oxysterols in Age-Related Disorders and Strategies to Alleviate Adverse Effects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:163-191. [PMID: 38036880 DOI: 10.1007/978-3-031-43883-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Oxysterols or cholesterol oxidation products are a class of molecules with the sterol moiety, derived from oxidative reaction of cholesterol through enzymatic and non-enzymatic processes. They are widely reported in animal-origin foods and prove significant involvement in the regulation of cholesterol homeostasis, lipid transport, cellular signaling, and other physiological processes. Reports of oxysterol-mediated cytotoxicity are in abundance and thus consequently implicated in several age-related and lifestyle disorders such as cardiovascular diseases, bone disorders, pancreatic disorders, age-related macular degeneration, cataract, neurodegenerative disorders such as Alzheimer's and Parkinson's disease, and some types of cancers. In this chapter, we attempt to review a selection of physiologically relevant oxysterols, with a focus on their formation, properties, and roles in health and disease, while also delving into the potential of natural and synthetic molecules along with bacterial enzymes for mitigating oxysterol-mediated cell damage.
Collapse
Affiliation(s)
- Shubhrima Ghosh
- Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Imen Ghzaiel
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
- Faculty of Medicine, Laboratory 'Nutrition, Functional Food and Vascular Health' (LR12ES05), University of Monastir, Monastir, Tunisia
| | - Anne Vejux
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Steve Meaney
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin 7, Ireland
| | - Sagnik Nag
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Gérard Lizard
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Garima Tripathi
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Falal Naez
- Department of Microbiology, Vijaygarh Jyotish Ray College, University of Calcutta, Kolkata, India
| | - Srijita Paul
- Department of Microbiology, Gurudas College, Kolkata, West Bengal, India
| |
Collapse
|
6
|
Ren C, Chen W, Chen J, Mao C, Liao C, Liu J. The fusion of keratinized epithelium, an indication of early implant placement in the aesthetic area: an animal study. BMC Oral Health 2023; 23:1016. [PMID: 38115040 PMCID: PMC10729501 DOI: 10.1186/s12903-023-03755-9] [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: 10/20/2023] [Accepted: 12/10/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND In the period of the early implant placement, the socket is mainly occupied by provisional matrix (PM). Keratinized epithelium (KE) is critical for primary wound closure. Although both KE and PM are important, the detailed relationship among migrating KE, PM formation and indication of the early implant placement is still unclear. OBJECTIVE This research aimed to locate a healing stage of KE with highest osteogenic PM formation after tooth extraction, which could be treated as the optimal time point for early implant placement. MATERIAL AND METHODS Mice were sacrificed on days 1, 2, 3, 4 and 6 after incisor extraction. Clinical, histological, and immunohistochemical evaluations of the extraction sockets were performed, and statistical analyses were conducted. We then inserted implants into the PM with the greatest bioactivity and observed its osseointegration pattern for 3, 10, 17 and 30 days. RESULT When KE fusion was reached, sockets were dominated by PM with the greatest expression of osteocalcin (OC, P < 0.05) and high levels of CD34 and Runx2. OC and Runx2 expression were positively correlated with KE coverage (P < 0.05). When the implant was inserted at 4 days' healing, the PM maintained its osteogenic ability, and osseointegration proceeded perfectly. CONCLUSION The migration of KE was correlated with the formation of highly osteogenic and angiogenic PM. And the fusion of KE could be treated as an indication for early implant placement.
Collapse
Affiliation(s)
- Chengyan Ren
- Department of Oral and Maxillofacial Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Weihui Chen
- Department of Oral and Maxillofacial Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Jiangping Chen
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanqing Mao
- Department of Oral and Maxillofacial Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Caiyu Liao
- Department of Oral and Maxillofacial Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jianan Liu
- Department of Oral and Maxillofacial Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
7
|
González-Salvatierra S, García-Fontana C, Lacal J, Andújar-Vera F, Martínez-Heredia L, Sanabria-de la Torre R, Ferrer-Millán M, Moratalla-Aranda E, Muñoz-Torres M, García-Fontana B. Cardioprotective function of sclerostin by reducing calcium deposition, proliferation, and apoptosis in human vascular smooth muscle cells. Cardiovasc Diabetol 2023; 22:301. [PMID: 37919715 PMCID: PMC10623848 DOI: 10.1186/s12933-023-02043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Sclerostin is an inhibitor of the Wnt/b-catenin pathway, which regulates bone formation, and can be expressed in vascular smooth muscle cells (VSMCs). Type 2 diabetes (T2D) is associated with an increased risk of cardiovascular disease (CVD) and increased serum and tissue expression of sclerostin. However, whether the role of sclerostin is detrimental or protective in the development of CVD is unknown. Therefore, our aims are to determine the level of sclerostin in T2D patients with/without CVD and in controls, both at serum and vascular tissue, and to analyze the role of sclerostin in VSMCs under calcified environments. METHODS Cross-sectional study including 121 controls and 139 T2D patients with/without CVD (48/91). Sclerostin levels in serum were determined by ELISA, and sclerostin expression was analyzed by RT-qPCR and immunohistochemistry in calcified and non-calcified artery of lower limb from T2D patients (n = 7) and controls (n = 3). In vitro experiments were performed in VSMCs (mock and sclerostin overexpression) under calcifying conditions analyzing the sclerostin function by determination of calcium and phosphate concentrations, and quantification of calcium deposits by Alizarin Red. Proliferation and apoptosis were analyzed by MTT assay and flow cytometry, respectively. The regulation of the expression of genes involved in bone metabolism was determined by RT-qPCR. RESULTS A significant increase in serum sclerostin levels in T2D patients with CVD compared to T2D patients without CVD and controls (p < 0.001) was observed. Moreover, higher circulating sclerostin levels were independently associated with CVD in T2D patients. Increased sclerostin expression was observed in calcified arteries of T2D patients compared to non-calcified arteries of controls (p = 0.003). In vitro experiments using VSMCs under calcified conditions, revealed that sclerostin overexpression reduced intracellular calcium (p = 0.001), calcium deposits (p < 0.001), cell proliferation (p < 0.001) and promoted cell survival (p = 0.015). Furthermore, sclerostin overexpression exhibited up-regulation of ALPL (p = 0.009), RUNX2 (p = 0.001) and COX2 (p = 0.003) and down-regulation of inflammatory genes, such as, IL1β (p = 0.005), IL6 (p = 0.001) and IL8 (p = 0.003). CONCLUSIONS Sclerostin could play a protective role in the development of atherosclerosis in T2D patients by reducing calcium deposits, decreasing proliferation and inflammation, and promoting cell survival in VSMCs under calcifying conditions. Therefore, considering the bone-vascular axis, treatment with anti-sclerostin for bone disease should be used with caution.
Collapse
Affiliation(s)
- Sheila González-Salvatierra
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology II, University of Granada, Granada, 18071, Spain
| | - Cristina García-Fontana
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain.
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, Granada, 18016, Spain.
- Institute of Health Carlos III, CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, 28029, Spain.
| | - Jesus Lacal
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Francisco Andújar-Vera
- Bioinformatic Research Service, Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Department of Computer Science and Artificial Intelligence, University of Granada, Granada, 18071, Spain
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI Institute), Granada, 18014, Spain
| | | | - Raquel Sanabria-de la Torre
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Department of Biochemistry, Molecular Biology III and Immunology, University of Granada, Granada, 18071, Spain
| | - María Ferrer-Millán
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
| | - Enrique Moratalla-Aranda
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Nuclear Medicine Unit, University Hospital Clínico San Cecilio, Granada, 18016, Spain
| | - Manuel Muñoz-Torres
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain.
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, Granada, 18016, Spain.
- Institute of Health Carlos III, CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, 28029, Spain.
- Department of Medicine, University of Granada, Granada, 18016, Spain.
| | - Beatriz García-Fontana
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, 18012, Spain
- Institute of Health Carlos III, CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, 28029, Spain
- Department of Cell Biology, University of Granada, Granada, 18016, Spain
| |
Collapse
|
8
|
Lu W, Sun C, Hou J. Predicting key gene related to immune infiltration and myofibroblast-like valve interstitial cells in patients with calcified aortic valve disease based on bioinformatics analysis. J Thorac Dis 2023; 15:3726-3740. [PMID: 37559614 PMCID: PMC10407485 DOI: 10.21037/jtd-23-72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 06/09/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Calcified aortic valve disease (CAVD) is the most prevalent valvular disease that can be treated only through valve replacement. We aimed to explore potential biomarkers and the role of immune cell infiltration in CAVD progression through bioinformatics analysis. METHODS Differentially ex-pressed genes (DEGs) were screened out based on three microarray datasets: GSE12644, GSE51472 and GSE83453. Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed to evaluate gene expression differences. Machine learning algorithms and DEGs were used to screen key gene. We used CIBERSORT to evaluate the immune cell infiltration of CAVD and evaluated the correlation between the biomarkers and infiltrating immune cells. We also compared bioinformatics analysis results with the valve interstitial cells (VICs) gene expression in single-cell RNA sequencing. RESULTS Collagen triple helix repeat containing 1 (CTHRC1) was identified as the key gene of CAVD. We identified a cell subtype valve interstitial cells-fibroblast, which was closely associated with fibro-calcific progress of aortic valve. CTHRC1 highly expressed in the VIC subpopulation. Immune infiltration analysis demonstrated that mast cells, B cells, dendritic cells and eosinophils were involved in pathogenesis of CAVD. Correlation analysis demonstrated that CTHRC1 was correlated with mast cells mostly. CONCLUSIONS In summary, the study suggested that CTHRC1 was a key gene of CAVD and CTHRC1 might participate in the potential molecular pathways involved in the connection between infiltrating immune cells and myofibroblast phenotype VICs.
Collapse
Affiliation(s)
- Wenyuan Lu
- Cardiac Surgery Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Sun
- Cardiac Surgery Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianfeng Hou
- Cardiac Surgery Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Leibowitz D, Yoshida Y, Jin Z, Nakanishi K, Mannina C, Elkind MSV, Rundek T, Homma S, Sacco RL, Di Tullio MR. Factors associated with the progression of aortic valve calcification in older adults. Int J Cardiol 2023; 381:76-80. [PMID: 37030403 PMCID: PMC10161393 DOI: 10.1016/j.ijcard.2023.03.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND Aortic valve calcification (AVC) is a common valvular abnormality that predisposes to stenosis; AVC progression and factors associated with it remain unclear. We investigated the association of clinical factors and serum biomarkers with AVC progression in a population-based cohort of older adults. METHODS Participants enrolled in both the Cardiovascular Abnormalities and Brain Lesion study (CABL; years 2005-2010) and the Subclinical Atrial Fibrillation And Risk of Ischemic Stroke study (SAFARIS;2014-2019) represent the study cohort. AVC was defined as bright dense echoes >1 mm in size on ≥1 cusps; each cusp was graded on a scale of 0 (normal) to 3 (severe calcification) at baseline and follow up. Serum biomarkers were measured at the time of follow-up assessment. RESULTS 373 participants (mean 68.1 ± 7.6 years of age, 146 M/ 227F) were included. 139 (37%) had AVC progression;93 (25%) had mild progression (1 grade), and 46 (12%) had moderate-severe progression (≥2 grades). The only significant clinical predictor of any progression was the use of anti-hypertensive medication which was associated with older age, higher BMI and more frequent hypertension, diabetes and hyperlipidemia. In multivariable analysis including biomarkers, transforming growth factor beta 1 (TGF-β1) was significantly associated with both all and moderate-severe AVC progression. CONCLUSIONS A significant number of elderly subjects with AVC show progression of their valve disease; individual vascular risk factors are not associated with AVC progression, although a combined effect may exist. Higher levels of TGF-β1 are observed in individuals with AVC progression.
Collapse
Affiliation(s)
- David Leibowitz
- Heart Institute, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuriko Yoshida
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Zhezhen Jin
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Koki Nakanishi
- Department of Cardiovascular Medicine, The University of Tokyo, Tokyo, Japan
| | - Carlo Mannina
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Mitchell S V Elkind
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Tatjana Rundek
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, FL, USA; Department of Public Health Sciences, Miller School of Medicine, University of Miami, FL, USA; Clinical and Translational Science Institute, Miller School of Medicine, University of Miami, FL, USA
| | - Shunichi Homma
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ralph L Sacco
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, FL, USA; Department of Public Health Sciences, Miller School of Medicine, University of Miami, FL, USA; Clinical and Translational Science Institute, Miller School of Medicine, University of Miami, FL, USA
| | - Marco R Di Tullio
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
10
|
Neels JG, Leftheriotis G, Chinetti G. Atherosclerosis Calcification: Focus on Lipoproteins. Metabolites 2023; 13:metabo13030457. [PMID: 36984897 PMCID: PMC10056669 DOI: 10.3390/metabo13030457] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipids in the vessel wall, leading to the formation of an atheroma and eventually to the development of vascular calcification (VC). Lipoproteins play a central role in the development of atherosclerosis and VC. Both low- and very low-density lipoproteins (LDL and VLDL) and lipoprotein (a) (Lp(a)) stimulate, while high-density lipoproteins (HDL) reduce VC. Apolipoproteins, the protein component of lipoproteins, influence the development of VC in multiple ways. Apolipoprotein AI (apoAI), the main protein component of HDL, has anti-calcific properties, while apoB and apoCIII, the main protein components of LDL and VLDL, respectively, promote VC. The role of lipoproteins in VC is also related to their metabolism and modifications. Oxidized LDL (OxLDL) are more pro-calcific than native LDL. Oxidation also converts HDL from anti- to pro-calcific. Additionally, enzymes such as autotaxin (ATX) and proprotein convertase subtilisin/kexin type 9 (PCSK9), involved in lipoprotein metabolism, have a stimulatory role in VC. In summary, a better understanding of the mechanisms by which lipoproteins and apolipoproteins contribute to VC will be crucial in the development of effective preventive and therapeutic strategies for VC and its associated cardiovascular disease.
Collapse
Affiliation(s)
- Jaap G Neels
- Université Côte d'Azur, INSERM, C3M, 06200 Nice, France
| | | | - Giulia Chinetti
- Université Côte d'Azur, CHU, INSERM, C3M, 06200 Nice, France
| |
Collapse
|
11
|
Molnár AÁ, Pásztor D, Merkely B. Cellular Senescence, Aging and Non-Aging Processes in Calcified Aortic Valve Stenosis: From Bench-Side to Bedside. Cells 2022; 11:cells11213389. [PMID: 36359785 PMCID: PMC9659237 DOI: 10.3390/cells11213389] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/30/2022] Open
Abstract
Aortic valve stenosis (AS) is the most common valvular heart disease. The incidence of AS increases with age, however, a significant proportion of elderly people have no significant AS, indicating that both aging and nonaging pathways are involved in the pathomechanism of AS. Age-related and stress-induced cellular senescence accompanied by further active processes represent the key elements of AS pathomechanism. The early stage of aortic valve degeneration involves dysfunction and disruption of the valvular endothelium due to cellular senescence and mechanical stress on blood flow. These cells are replaced by circulating progenitor cells, but in an age-dependent decelerating manner. When endothelial denudation is no longer replaced by progenitor cells, the path opens for focal lipid deposition, initiating subsequent oxidation, inflammation and micromineralisation. Later stages of AS feature a complex active process with extracellular matrix remodeling, fibrosis and calcification. Echocardiography is the gold standard method for diagnosing aortic valve disease, although computed tomography and cardiac magnetic resonance are useful additional imaging methods. To date, no medical treatment has been proven to halt the progression of AS. Elucidation of differences and similarities between vascular and valvular calcification pathomechanisms may help to find effective medical therapy and reduce the increasing health burden of the disease.
Collapse
|
12
|
Chen L, Liu H, Sun C, Pei J, Li J, Li Y, Wei K, Wang X, Wang P, Li F, Gai S, Zhao Y, Zheng Z. A Novel LncRNA SNHG3 Promotes Osteoblast Differentiation Through BMP2 Upregulation in Aortic Valve Calcification. JACC Basic Transl Sci 2022; 7:899-914. [PMID: 36317131 PMCID: PMC9617132 DOI: 10.1016/j.jacbts.2022.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022]
Abstract
The long noncoding RNA SNHG3 was upregulated in the leaflets of both patients and mice with calcific aortic valve disease. SNHG3 can associate with EZH2 in the nucleus of hVICs to epigenetically upregulate BMP2, a key mediator of calcification. SNHG3 promoted osteoblast differentiation of hVICs via upregulation of the BMP2 pathway. SNHG3 silencing significantly ameliorated aortic valve calcification in experimental animals, providing a novel therapeutic target for CAVD.
Based on high-throughput transcriptomic sequencing, SNHG3 was among the most highly expressed long noncoding RNAs in calcific aortic valve disease. SNHG3 upregulation was verified in human and mouse calcified aortic valves. Moreover, in vivo and in vitro studies showed SNHG3 silencing markedly ameliorated aortic valve calcification. In-depth functional assays showed SNHG3 physically interacted with polycomb repressive complex 2 to suppress the H3K27 trimethylation BMP2 locus, which in turn activated BMP2 expression and signaling pathways. Taken together, SNHG3 promoted aortic valve calcification by upregulating BMP2, which might be a novel therapeutic target in human calcific aortic valve disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Zhe Zheng
- Address for correspondence: Dr Zhe Zheng, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, China and Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Beilishi Road, Xicheng District, Beijing, PR China.
| |
Collapse
|
13
|
Leifheit-Nestler M, Vogt I, Haffner D, Richter B. Phosphate Is a Cardiovascular Toxin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:107-134. [DOI: 10.1007/978-3-030-91623-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
14
|
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
|
15
|
Waring OJ, Skenteris NT, Biessen EAL, Donners MMPC. Two-faced Janus: The dual role of macrophages in atherosclerotic calcification. Cardiovasc Res 2021; 118:2768-2777. [PMID: 34550346 PMCID: PMC9586561 DOI: 10.1093/cvr/cvab301] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/20/2021] [Indexed: 12/19/2022] Open
Abstract
Calcification is an independent predictor of atherosclerosis-related cardiovascular events. Microcalcification is linked to inflamed, unstable lesions, in comparison to the fibrotic stable plaque phenotype generally associated with advanced calcification. This paradox relates to recognition that calcification presents in a wide spectrum of manifestations that differentially impact plaque’s fate. Macrophages, the main inflammatory cells in atherosclerotic plaque, have a multifaceted role in disease progression. They crucially control the mineralization process, from microcalcification to the osteoid metaplasia of bone-like tissue. It is a bilateral interaction that weighs heavily on the overall plaque fate but remains rather unexplored. This review highlights current knowledge about macrophage phenotypic changes in relation to and interaction with the calcifying environment. On the one hand, macrophage-led inflammation kickstarts microcalcification through a multitude of interlinked mechanisms, which in turn stimulates phenotypic changes in vascular cell types to drive microcalcification. Macrophages may also modulate the expression/activity of calcification inhibitors and inducers, or eliminate hydroxyapatite nucleation points. Contrarily, direct exposure of macrophages to an early calcifying milieu impacts macrophage phenotype, with repercussions for plaque progression and/or stability. Macrophages surrounding macrocalcification deposits show a more reparative phenotype, modulating extracellular matrix, and expressing osteoclast genes. This phenotypic shift favours gradual displacement of the pro-inflammatory hubs; the lipid necrotic core, by macrocalcification. Parallels to bone metabolism may explain many of these changes to macrophage phenotype, with advanced calcification able to show homeostatic osteoid metaplasia. As the targeted treatment of vascular calcification developing in atherosclerosis is thus far severely lacking, it is crucial to better understand its mechanisms of development.
Collapse
Affiliation(s)
- O J Waring
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - N T Skenteris
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands.,Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - E A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands.,Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, German
| | - M M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
16
|
Masbuchin AN, Rohman MS, Liu PY. Role of Glycosylation in Vascular Calcification. Int J Mol Sci 2021; 22:9829. [PMID: 34575990 PMCID: PMC8469761 DOI: 10.3390/ijms22189829] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Glycosylation is an important step in post-translational protein modification. Altered glycosylation results in an abnormality that causes diseases such as malignancy and cardiovascular diseases. Recent emerging evidence highlights the importance of glycosylation in vascular calcification. Two major types of glycosylation, N-glycosylation and O-glycosylation, are involved in vascular calcification. Other glycosylation mechanisms, which polymerize the glycosaminoglycan (GAG) chain onto protein, resulting in proteoglycan (PG), also have an impact on vascular calcification. This paper discusses the role of glycosylation in vascular calcification.
Collapse
Affiliation(s)
- Ainun Nizar Masbuchin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan;
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Brawijaya, Malang 65111, Indonesia;
| | - Mohammad Saifur Rohman
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Brawijaya, Malang 65111, Indonesia;
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan;
- Division of Cardiology, Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| |
Collapse
|
17
|
Xu B, Kocyigit D, Wang TKM, Tan CD, Rodriguez ER, Pettersson GB, Unai S, Griffin BP. Mitral annular calcification and valvular dysfunction: multimodality imaging evaluation, grading, and management. Eur Heart J Cardiovasc Imaging 2021; 23:e111-e122. [PMID: 34591959 DOI: 10.1093/ehjci/jeab185] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/02/2021] [Indexed: 11/12/2022] Open
Abstract
Mitral annular calcification (MAC) refers to calcium deposition in the fibrous skeleton of the mitral valve. It has many cardiovascular associations, including mitral valve dysfunction, elevated cardiovascular risk, arrhythmias, and endocarditis. Echocardiography conventionally is the first-line imaging modality for anatomic assessment, and evaluation of mitral valve function. Cardiac computed tomography (CT) has demonstrated importance as an imaging modality for the evaluation and planning of related procedures. It also holds promise in quantitative grading of MAC. Currently, there is no universally accepted definition or classification system of MAC severity. We review the multimodality imaging evaluation of MAC and associated valvular dysfunction and propose a novel classification system based on qualitative and quantitative measurements derived from echocardiography and cardiac CT.
Collapse
Affiliation(s)
- Bo Xu
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J1-5, Cleveland, OH 44195, USA
| | - Duygu Kocyigit
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J1-5, Cleveland, OH 44195, USA
| | - Tom Kai Ming Wang
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J1-5, Cleveland, OH 44195, USA
| | - Carmela D Tan
- Department of Cardiovascular Anatomical Pathology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - E Rene Rodriguez
- Department of Cardiovascular Anatomical Pathology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gösta B Pettersson
- Department of Thoracic and Cardiovascular Surgery, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shinya Unai
- Department of Thoracic and Cardiovascular Surgery, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Brian P Griffin
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J1-5, Cleveland, OH 44195, USA
| |
Collapse
|
18
|
Radvar E, Griffanti G, Tsolaki E, Bertazzo S, Nazhat SN, Addison O, Mata A, Shanahan CM, Elsharkawy S. Engineered In vitro Models for Pathological Calcification: Routes Toward Mechanistic Understanding. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Elham Radvar
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| | - Gabriele Griffanti
- Department of Mining and Materials Engineering Faculty of Engineering McGill University Montreal QC H3A 0C5 Canada
| | - Elena Tsolaki
- Department of Medical Physics and Biomedical Engineering University College London London WC1E 6BT UK
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering University College London London WC1E 6BT UK
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering Faculty of Engineering McGill University Montreal QC H3A 0C5 Canada
| | - Owen Addison
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| | - Alvaro Mata
- School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
| | - Catherine M. Shanahan
- BHF Centre of Research Excellence Cardiovascular Division James Black Centre King's College London London SE1 1UL UK
| | - Sherif Elsharkawy
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| |
Collapse
|
19
|
Lin S, Li X, Zhang J, Zhang Y. Omentin-1: Protective impact on ischemic stroke via ameliorating atherosclerosis. Clin Chim Acta 2021; 517:31-40. [PMID: 33607071 DOI: 10.1016/j.cca.2021.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/26/2022]
Abstract
Omentin-1, a newly identified adipokine, has recently been revealed as a novel biomarker for ischemic stroke (IS). Low circulating omentin-1 levels could indicate a high risk of IS, and elevated omentin-1 levels exert a favorable impact on cerebral ischemia. Furthermore, omentin-1 has anti-atherosclerotic, anti-inflammatory, and cardiovascular protective capabilities through the intracellular Akt/AMP-activated protein kinase (AMPK)/ nuclear factor-κB (NF-κB) and certain protein kinase (ERK, JNK, and p38) signaling pathways. Omentin-1 also alleviates endothelial cell dysfunction, improves revascularization via the Akt-endothelial nitric-oxide synthase (eNOS) regulatory axis, promotes endothelium-dependent vasodilation through endothelium-derived NO in an eNOS fashion, and inhibits VSMC proliferation by means of AMPK/ERK signaling pathways, VSMC migration via inactivation of the NADPH oxidase (NOX)/ROS/p38/HSP27 pathways and artery calcification via the PI3K-Akt pathway. These findings indicate that omentin-1 may be a negative mediator of IS. Pharmacologically, several lines of clinical evidence indicate that metformin and statins could elevate omentin-1 levels, although the specific mechanism has not been precisely delineated until now. This study is the first to summarize the comprehensive mechanisms between omentin-1 and atherosclerosis and to review the shielding effect of omentin-1 on IS. We shed light on omentin-1 as a novel therapeutic target for combating IS.
Collapse
Affiliation(s)
- Shiyi Lin
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China; School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Li
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiabei Zhang
- School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Yuyang Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
20
|
Vidavsky N, Kunitake JAMR, Estroff LA. Multiple Pathways for Pathological Calcification in the Human Body. Adv Healthc Mater 2021; 10:e2001271. [PMID: 33274854 PMCID: PMC8724004 DOI: 10.1002/adhm.202001271] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/16/2020] [Indexed: 12/12/2022]
Abstract
Biomineralization of skeletal components (e.g., bone and teeth) is generally accepted to occur under strict cellular regulation, leading to mineral-organic composites with hierarchical structures and properties optimized for their designated function. Such cellular regulation includes promoting mineralization at desired sites as well as inhibiting mineralization in soft tissues and other undesirable locations. In contrast, pathological mineralization, with potentially harmful health effects, can occur as a result of tissue or metabolic abnormalities, disease, or implantation of certain biomaterials. This progress report defines mineralization pathway components and identifies the commonalities (and differences) between physiological (e.g., bone remodeling) and pathological calcification formation pathways, based, in part, upon the extent of cellular control within the system. These concepts are discussed in representative examples of calcium phosphate-based pathological mineralization in cancer (breast, thyroid, ovarian, and meningioma) and in cardiovascular disease. In-depth mechanistic understanding of pathological mineralization requires utilizing state-of-the-art materials science imaging and characterization techniques, focusing not only on the final deposits, but also on the earlier stages of crystal nucleation, growth, and aggregation. Such mechanistic understanding will further enable the use of pathological calcifications in diagnosis and prognosis, as well as possibly provide insights into preventative treatments for detrimental mineralization in disease.
Collapse
Affiliation(s)
- Netta Vidavsky
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Jennie A M R Kunitake
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY, 14853, USA
| |
Collapse
|
21
|
Lee HY, Lim S, Park S. Role of Inflammation in Arterial Calcification. Korean Circ J 2021; 51:114-125. [PMID: 33525066 PMCID: PMC7853899 DOI: 10.4070/kcj.2020.0517] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 01/11/2023] Open
Abstract
Arterial calcification, characterized by calcium phosphate deposition in the arteries, can be divided into intimal calcification and medial calcification. The former is the predominant form of calcification in coronary artery plaques; the latter mostly affects peripheral arteries and aortas. Both forms of arterial calcification have strong correlations with adverse cardiovascular events. Intimal microcalcification is associated with increased risk of plaque disruption while the degree of burden of coronary calcification, measured by coronary calcium score, is a marker of overall plaque burden. Continuous research on vascular calcification has been performed during the past few decades, and several cellular and molecular mechanisms and therapeutic targets were identified. However, despite clinical trials to evaluate the efficacy of drug therapies to treat vascular calcification, none have been shown to have efficacy until the present. Therefore, more extensive research is necessary to develop appropriate therapeutic strategies based on a thorough understanding of vascular calcification. In this review, we mainly focus on intimal calcification, namely the pathobiology of arterial calcification, and its clinical implications.
Collapse
Affiliation(s)
- Hae Young Lee
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, Seoul, Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Korea
| | - Sungha Park
- Division of Cardiology, Severance Cardiovascular Hospital and Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
22
|
Yang P, Troncone L, Augur ZM, Kim SSJ, McNeil ME, Yu PB. The role of bone morphogenetic protein signaling in vascular calcification. Bone 2020; 141:115542. [PMID: 32736145 PMCID: PMC8185454 DOI: 10.1016/j.bone.2020.115542] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/02/2020] [Accepted: 07/04/2020] [Indexed: 01/10/2023]
Abstract
Vascular calcification is associated with atherosclerosis, chronic kidney disease, and diabetes, and results from processes resembling endochondral or intramembranous ossification, or from processes that are distinct from ossification. Bone morphogenetic proteins (BMP), as well as other ligands, receptors, and regulators of the transforming growth factor beta (TGFβ) family regulate vascular and valvular calcification by modulating the phenotypic plasticity of multipotent progenitor lineages associated with the vasculature or valves. While osteogenic ligands BMP2 and BMP4 appear to be both markers and drivers of vascular calcification, particularly in atherosclerosis, BMP7 may serve to protect against calcification in chronic kidney disease. BMP signaling regulators such as matrix Gla protein and BMP-binding endothelial regulator protein (BMPER) play protective roles in vascular calcification. The effects of BMP signaling molecules in vascular calcification are context-dependent, tissue-dependent, and cell-type specific. Here we review the current knowledge on mechanisms by which BMP signaling regulates vascular calcification and the potential therapeutic implications.
Collapse
Affiliation(s)
- Peiran Yang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Luca Troncone
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zachary M Augur
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stephanie S J Kim
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Megan E McNeil
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Cocci A, Di Maida F, Russo GI, Capogrosso P, Francesco L, Rizzo M, Di Mauro M, Salonia A, Cito G, Falcone M, Romano A, Polloni G, Martinez-Salamanca JI, Fernández-Pascual E, Minervini A, Mondaini N. Efficacy of Collagenase Clostridium histolyticum (Xiapex ®) in Patients with the Acute Phase of Peyronie's Disease. Clin Drug Investig 2020; 40:583-588. [PMID: 32342279 DOI: 10.1007/s40261-020-00916-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND OBJECTIVE: Plaque formation ordinarily takes place in the acute phase of Peyronie's disease. There is no unanimous consent regarding the management of the acute phase of Peyronie's disease. The objective of this study was to evaluate the advantages of using a single intralesional injection of collagenase Clostridium histolyticum in patients with the active phase of Peyronie's disease and to assess its effect on disease progression by reducing penile curvature and ameliorating pain during sexual intercourse. METHODS Sexually active men aged older than 18 years with the acute phase of Peyronie's disease were enrolled. All patients received treatment with a single intralesional injection of collagenase Clostridium histolyticum. The primary outcome of the study was the change in penile curvature after treatment while secondary outcomes were the change in sexual function (International Index of Erectile Function [IIEF-5]) and in the Peyronie Disease Questionnaire (PDQ) and its sub-scores, PDQ-PS (psychological symptoms), PDQ-PP (penile pain) and PDQ-BD (bother disease). RESULTS Overall, 74 patients were enrolled. Mean penile curvature at baseline was 41.1° ± 12.2°. The mean changes before and at the 3-month evaluation in terms of penile curvature, Visual Analog Scale score at rest, and Visual Analog Scale score during intercourse were - 19.3 ± 8.4 (p < 0.0001), - 0.8 ± 1.1 (p < 0.0001) and - 3.8 ± 0.9 (p < 0.0001) with the benefit persisting also after 6 months. Moreover, improvements of mean IIEF-5 score (1.1 ± 0.9, p = 0.03; 0.9 ± 0.5, p = 0.02), PDQ-PS (- 2.7 ± 2.2; - 2.5 ± 2.0, p = 0.01), PDQ-PP (- 1.2 ± 1.6; - 1.1 ± 1.2, p = 0.02) and PDQ-BD (- 3.8 ± 3.4; - 3.5 ± 3.1, p = 0.001) were observed 3 and 6 months after the end of treatment, respectively. At the multivariable regression analysis, the time since disease onset (modelled with non-linear terms) and baseline curvature were independently associated with the degree of curvature improvement (coefficient: 0.30; 95% confidence interval 0.16-0.44) after a single intralesional injection (all p < 0.03). CONCLUSIONS Although intralesional therapy with collagenase Clostridium histolyticum is not yet indicated for the acute phase of Peyronie's disease, these preliminary results suggest the effectiveness of this minimally invasive option by improving penile curvature and IIEF-5 and PDQ scores.
Collapse
Affiliation(s)
- Andrea Cocci
- Department of Urology, Careggi Hospital, University of Florence, Viale S. Luca, 50134, Florence, FI, Italy
| | - Fabrizio Di Maida
- Department of Urology, Careggi Hospital, University of Florence, Viale S. Luca, 50134, Florence, FI, Italy.
| | - Giorgio Ivan Russo
- Urology Section, Department of Surgery, University of Catania, Catania, Italy
| | - Paolo Capogrosso
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Lotti Francesco
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Michele Rizzo
- Department of Urology, University of Trieste, Trieste, Italy
| | - Marina Di Mauro
- Urology Section, Department of Surgery, University of Catania, Catania, Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Gianmartin Cito
- Department of Urology, Careggi Hospital, University of Florence, Viale S. Luca, 50134, Florence, FI, Italy
| | - Marco Falcone
- Urology Clinic, A.O.U. Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Andrea Romano
- Department of Urology, Careggi Hospital, University of Florence, Viale S. Luca, 50134, Florence, FI, Italy
| | | | | | - Esaù Fernández-Pascual
- Department of Urology, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Andrea Minervini
- Department of Urology, Careggi Hospital, University of Florence, Viale S. Luca, 50134, Florence, FI, Italy
| | | |
Collapse
|
24
|
Fedintsev A, Moskalev A. Stochastic non-enzymatic modification of long-lived macromolecules - A missing hallmark of aging. Ageing Res Rev 2020; 62:101097. [PMID: 32540391 DOI: 10.1016/j.arr.2020.101097] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/05/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Damage accumulation in long-living macromolecules (especially extracellular matrix (ECM) proteins, nuclear pore complex (NPC) proteins, and histones) is a missing hallmark of aging. Stochastic non-enzymatic modifications of ECM trigger cellular senescence as well as many other hallmarks of aging affect organ barriers integrity and drive tissue fibrosis. The importance of it for aging makes it a key target for interventions. The most promising of them can be AGE inhibitors (chelators, O-acetyl group or transglycating activity compounds, amadorins and amadoriases), glucosepane breakers, stimulators of elastogenesis, and RAGE antagonists.
Collapse
Affiliation(s)
- Alexander Fedintsev
- Institute of Biology of FRC of Komi Scientific Center, Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Alexey Moskalev
- Institute of Biology of FRC of Komi Scientific Center, Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia.
| |
Collapse
|
25
|
Zhang Y, Qian H, Wu B, You S, Wu S, Lu S, Wang P, Cao L, Zhang N, Sun Y. E3 Ubiquitin ligase NEDD4 family‑regulatory network in cardiovascular disease. Int J Biol Sci 2020; 16:2727-2740. [PMID: 33110392 PMCID: PMC7586430 DOI: 10.7150/ijbs.48437] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022] Open
Abstract
Protein ubiquitination represents a critical modification occurring after translation. E3 ligase catalyzes the covalent binding of ubiquitin to the protein substrate, which could be degraded. Ubiquitination as an important protein post-translational modification is closely related to cardiovascular disease. The NEDD4 family, belonging to HECT class of E3 ubiquitin ligases can recognize different substrate proteins, including PTEN, ENaC, Nav1.5, SMAD2, PARP1, Septin4, ALK1, SERCA2a, TGFβR3 and so on, via the WW domain to catalyze ubiquitination, thus participating in multiple cardiovascular-related disease such as hypertension, arrhythmia, myocardial infarction, heart failure, cardiotoxicity, cardiac hypertrophy, myocardial fibrosis, cardiac remodeling, atherosclerosis, pulmonary hypertension and heart valve disease. However, there is currently no review comprehensively clarifying the important role of NEDD4 family proteins in the cardiovascular system. Therefore, the present review summarized recent studies about NEDD4 family members in cardiovascular disease, providing novel insights into the prevention and treatment of cardiovascular disease. In addition, assessing transgenic animals and performing gene silencing would further identify the ubiquitination targets of NEDD4. NEDD4 quantification in clinical samples would also constitute an important method for determining NEDD4 significance in cardiovascular disease.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Hao Qian
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shaojun Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Saien Lu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Pingyuan Wang
- Staff scientist, Center for Molecular Medicine National Heart Lung and Blood Institute, National Institutes of Health, the United States
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
26
|
Dong Q, Chen Y, Liu W, Liu X, Chen A, Yang X, Li Y, Wang S, Fu M, Ou JS, Lu L, Yan J. 25-Hydroxycholesterol promotes vascular calcification via activation of endoplasmic reticulum stress. Eur J Pharmacol 2020; 880:173165. [PMID: 32423869 DOI: 10.1016/j.ejphar.2020.173165] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/23/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
Abstract
Vascular calcification is a highly regulated process similar to osteogenesis involving phenotypic change of vascular smooth muscle cells (VSMCs). 25-Hydroxycholesterol (25-HC), one of oxysterols synthesized by the enzyme cholesterol 25-hydroxylase, has been shown to promote bovine calcifying vascular cells (CVC) calcification. However, whether and how 25-HC regulates vascular calcification are not completely understood. In this study, in vitro and ex vivo models of vascular calcification were used to determine whether 25-HC regulates vascular calcification. Alizarin red staining and calcium content assay showed that 25-HC treatment promoted calcification of rat and human VSMCs in a dose-dependent manner. Similarly, ex vivo study further confirmed that 25-HC accelerated calcification of rat aortic rings. In addition, western blot analysis showed that 25-HC significantly up-regulated the expression of endoplasmic reticulum stress (ERS) signaling molecules including ATF4 and CHOP in VSMCs and flow cytometry analysis revealed that 25-HC increased apoptosis of VSMCs. Moreover, knockdown of CHOP by siRNA blocked 25-HC-induced mineral deposition in VSMCs. Collectively, this study for the first time demonstrates that 25-HC promotes vascular calcification via ATF4/CHOP signaling using in vitro and ex vivo models, suggesting that ERS is involved in the regulation of 25-HC-induced vascular calcification.
Collapse
MESH Headings
- Activating Transcription Factor 4/metabolism
- Animals
- Aorta, Thoracic/cytology
- Apoptosis/drug effects
- Calcium/metabolism
- Cells, Cultured
- Endoplasmic Reticulum Stress/drug effects
- Humans
- Hydroxycholesterols/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- RNA, Small Interfering/genetics
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Transcription Factor CHOP/genetics
- Vascular Calcification/chemically induced
- Vascular Calcification/metabolism
Collapse
Affiliation(s)
- Qianqian Dong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China
| | - Yanting Chen
- Department of Pathophysiolgy, Zhongshan School of Medicine, Sun Yat-Sen University, China
| | - Wantao Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China
| | - Xiaoyu Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China
| | - Xiulin Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China
| | - Yining Li
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China
| | - Siyi Wang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China
| | - Mingwei Fu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China
| | - Jing-Song Ou
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-Sen University, China
| | - Lihe Lu
- Department of Pathophysiolgy, Zhongshan School of Medicine, Sun Yat-Sen University, China.
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China.
| |
Collapse
|
27
|
Rogers MA, Aikawa E. Cardiovascular calcification: artificial intelligence and big data accelerate mechanistic discovery. Nat Rev Cardiol 2020; 16:261-274. [PMID: 30531869 DOI: 10.1038/s41569-018-0123-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular calcification is a health disorder with increasing prevalence and high morbidity and mortality. The only available therapeutic options for calcific vascular and valvular heart disease are invasive transcatheter procedures or surgeries that do not fully address the wide spectrum of these conditions; therefore, an urgent need exists for medical options. Cardiovascular calcification is an active process, which provides a potential opportunity for effective therapeutic targeting. Numerous biological processes are involved in calcific disease, including matrix remodelling, transcriptional regulation, mitochondrial dysfunction, oxidative stress, calcium and phosphate signalling, endoplasmic reticulum stress, lipid and mineral metabolism, autophagy, inflammation, apoptosis, loss of mineralization inhibition, impaired mineral resorption, cellular senescence and extracellular vesicles that act as precursors of microcalcification. Advances in molecular imaging and big data technology, including in multiomics and network medicine, and the integration of these approaches are helping to provide a more comprehensive map of human disease. In this Review, we discuss ectopic calcification processes in the cardiovascular system, with an emphasis on emerging mechanistic knowledge obtained through patient data and advances in imaging methods, experimental models and multiomics-generated big data. We also highlight the potential and challenges of artificial intelligence, machine learning and deep learning to integrate imaging and mechanistic data for drug discovery.
Collapse
Affiliation(s)
- Maximillian A Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Millar SA, John SG, McIntyre CW, Ralevic V, Anderson SI, O'Sullivan SE. An Investigation Into the Role of Osteocalcin in Human Arterial Smooth Muscle Cell Calcification. Front Endocrinol (Lausanne) 2020; 11:369. [PMID: 32587575 PMCID: PMC7298126 DOI: 10.3389/fendo.2020.00369] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022] Open
Abstract
Osteocalcin (OCN) is a bone-derived protein that is detected within human calcified vascular tissue. Calcification is particularly prevalent in chronic kidney disease (CKD) patients but the role of OCN in calcification, whether active or passive, has not been elucidated. Part 1: The relationship between OCN, CKD and vascular calcification was assessed in CKD patients (n = 28) and age-matched controls (n = 19). Part 2: in vitro, we analyzed whether addition of uncarboxylated osteocalcin (ucOCN) influenced the rate or extent of vascular smooth muscle cell (VSMC) calcification. Human aortic VSMCs were cultured in control media or mineralisation inducing media (MM) containing increased phosphate with or without ucOCN (10 or 30 ng/mL) for up to 21 days. Markers of osteogenic differentiation and calcification were determined [alkaline phosphatase (ALP) activity, total intracellular OCN, Runx2 expression, α-SMA expression, alizarin red calcium staining, and calcium quantification]. Part 1 results: In our human population, calcification was present (mean age 76 years), but no differences were detected between CKD patients and controls. Plasma total OCN was increased in CKD patients compared to controls (14 vs. 9 ng/mL; p < 0.05) and correlated to estimated glomerular filtration rate (p < 0.05), however no relationship was detected between total OCN and calcification. Part 2 results: in vitro, ALP activity, α-SMA expression and calcium concentrations were significantly increased in MM treated VSMCs at day 21, but no effect of ucOCN was observed. Cells treated with control media+ucOCN for 21 days did not show increases in ALP activity nor calcification. In summary, although plasma total OCN was increased in CKD patients, this study did not find a relationship between OCN and calcification in CKD and non-CKD patients, and found no in vitro evidence of an active role of ucOCN in vascular calcification as assessed over 21 days. ucOCN appears not to be a mediator of vascular calcification, but further investigation is warranted.
Collapse
Affiliation(s)
- Sophie A. Millar
- Division of Medical Sciences & Graduate Entry Medicine, School of Medicine, Royal Derby Hospital, University of Nottingham, Derby, United Kingdom
- *Correspondence: Sophie A. Millar
| | - Stephen G. John
- Department of Renal Medicine, Royal Derby Hospital, Derby, United Kingdom
| | - Christopher W. McIntyre
- Department of Renal Medicine, Royal Derby Hospital, Derby, United Kingdom
- London Health Sciences Centre, London, ON, Canada
| | - Vera Ralevic
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Susan I. Anderson
- Division of Medical Sciences & Graduate Entry Medicine, School of Medicine, Royal Derby Hospital, University of Nottingham, Derby, United Kingdom
| | - Saoirse E. O'Sullivan
- Division of Medical Sciences & Graduate Entry Medicine, School of Medicine, Royal Derby Hospital, University of Nottingham, Derby, United Kingdom
| |
Collapse
|
29
|
Grand Moursel L, van der Graaf LM, Bulk M, van Roon‐Mom WM, van der Weerd L. Osteopontin and phospho-SMAD2/3 are associated with calcification of vessels in D-CAA, an hereditary cerebral amyloid angiopathy. Brain Pathol 2019; 29:793-802. [PMID: 30868685 PMCID: PMC6850614 DOI: 10.1111/bpa.12721] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/06/2019] [Indexed: 01/06/2023] Open
Abstract
In severe forms of cerebral amyloid angiopathy (CAA) pathology, vascular calcification has been observed in the cerebral cortex, both in vivo on MRI and CT, and post-mortem using histopathology. However, the pathomechanisms leading to calcification of CAA-laden arteries are unknown. Therefore, we investigated the correlation between calcification of cortical arterioles and several potential modulators of vascular calcification using immunohistochemistry in a unique collection of brain material of patients with a hereditary form of CAA, namely hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D or D-CAA). We show a topographical association of osteopontin (OPN) and TGFβ signaling factor phospho-SMAD2/3 (pSMAD2/3) in calcified CAA vessel walls. OPN and pSMAD2/3 gradually accumulate in vessels prior to calcification. Moreover, we found that the vascular accumulation of Collagen 1 (Col1), OPN and pSMAD2/3 immunomarkers correlated with the CAA severity. This was independently of the vessel size, including capillaries in the most severe cases. We propose that calcification of CAA vessels in the observed HCHWA-D cases may be induced by extracellular OPN trapped in the fibrotic Col1 vessel wall, independently of the presence of vascular amyloid.
Collapse
Affiliation(s)
- Laure Grand Moursel
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Linda M. van der Graaf
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Marjolein Bulk
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| | | | - Louise van der Weerd
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
- Department of RadiologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
30
|
Creager MD, Hohl T, Hutcheson JD, Moss AJ, Schlotter F, Blaser MC, Park MA, Lee LH, Singh SA, Alcaide-Corral CJ, Tavares AAS, Newby DE, Kijewski MF, Aikawa M, Di Carli M, Dweck MR, Aikawa E. 18F-Fluoride Signal Amplification Identifies Microcalcifications Associated With Atherosclerotic Plaque Instability in Positron Emission Tomography/Computed Tomography Images. Circ Cardiovasc Imaging 2019; 12:e007835. [PMID: 30642216 DOI: 10.1161/circimaging.118.007835] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Microcalcifications in atherosclerotic plaques are destabilizing, predict adverse cardiovascular events, and are associated with increased morbidity and mortality.18F-fluoride positron emission tomography (PET)/computed tomography (CT) imaging has demonstrated promise as a useful clinical diagnostic tool in identifying high-risk plaques; however, there is confusion as to the underlying mechanism of signal amplification seen in PET-positive, CT-negative image regions. This study tested the hypothesis that 18F-fluoride PET/CT can identify early microcalcifications. METHODS 18F-fluoride signal amplification derived from microcalcifications was validated against near-infrared fluorescence molecular imaging and histology using an in vitro 3-dimensional hydrogel collagen platform, ex vivo human specimens, and a mouse model of atherosclerosis. RESULTS Microcalcification size correlated inversely with collagen concentration. The 18F-fluoride ligand bound to microcalcifications formed by calcifying vascular smooth muscle cell derived extracellular vesicles in the in vitro 3-dimensional collagen system and exhibited an increasing signal with an increase in collagen concentration (0.25 mg/mL collagen -33.8×102±12.4×102 counts per minute; 0.5 mg/mL collagen -67.7×102±37.4×102 counts per minute; P=0.0014), suggesting amplification of the PET signal by smaller microcalcifications. We further incubated human atherosclerotic endarterectomy specimens with clinically relevant concentrations of 18F-fluoride. The 18F-fluoride ligand labeled microcalcifications in PET-positive, CT-negative regions of explanted human specimens as evidenced by 18F-fluoride PET/CT imaging, near-infrared fluorescence, and histological analysis. Additionally, the 18F-fluoride ligand identified micro and macrocalcifications in atherosclerotic aortas obtained from low-density lipoprotein receptor-deficient mice. CONCLUSIONS Our results suggest that 18F-fluoride PET signal in PET-positive, CT-negative regions of human atherosclerotic plaques is the result of developing microcalcifications, and high surface area in regions of small microcalcifications may amplify PET signal.
Collapse
Affiliation(s)
- Michael D Creager
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Tobias Hohl
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | | | - Alastair J Moss
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - Florian Schlotter
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Mark C Blaser
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Mi-Ae Park
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.-A.P., M.F.K., M.D.C.)
| | - Lang Ho Lee
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Sasha A Singh
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.)
| | - Carlos J Alcaide-Corral
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - Adriana A S Tavares
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - David E Newby
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - Marie F Kijewski
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.-A.P., M.F.K., M.D.C.)
| | - Masanori Aikawa
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.).,Division of Cardiovascular Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.A., E.A.)
| | - Marcelo Di Carli
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.-A.P., M.F.K., M.D.C.)
| | - Marc R Dweck
- British Heart Foundation, Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (A.J.M., C.J.A.-C., A.A.S.T., D.E.N., M.R.D.)
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.D.C., T.H., F.S., M.C.B. L.H.L., S.A.S., M.A., E.A.).,Division of Cardiovascular Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (M.A., E.A.)
| |
Collapse
|
31
|
Abstract
Vascular calcification (VC) is strongly associated with all-cause mortality and is an independent predictor of cardiovascular events. Resulting from its complex, multifaceted nature, targeted treatments for VC have not yet been developed. Lipoproteins are well characterized in the pathogenesis of atherosclerotic plaques, leading to the development of plaque regressing therapeutics. Although their roles in plaque progression are well documented, their roles in VC, and calcification of a plaque, are not well understood. In this review, early in vitro data and clinical correlations suggest an inhibitory role for HDL (high-density lipoproteins) in VC, a stimulatory role for LDL (low-density lipoprotein) and VLDL (very low-density lipoprotein) and a potentially causal role for Lp(a) (lipoprotein [a]). Additionally, after treatment with a statin or PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitor, plaque calcification is observed to increase. With the notion that differing morphologies of plaque calcification associate with either a more stable or unstable plaque phenotype, uncovering the mechanisms of lipoprotein-artery wall interactions could produce targeted therapeutic options for VC.
Collapse
Affiliation(s)
- Emma J. Akers
- From the South Australian Health and Medical Research Institute, Adelaide, Australia (E.J.A.)
- The University of Adelaide, Australia (E.J.A.)
| | - Stephen J. Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Australia (S.J.N.)
| | - Belinda A. Di Bartolo
- The Kolling Institute of Medical Research, The University of Sydney, Australia (B.A.D.B.)
| |
Collapse
|
32
|
Babic M, Schuchardt M, Tölle M, van der Giet M. In times of tobacco-free nicotine consumption: The influence of nicotine on vascular calcification. Eur J Clin Invest 2019; 49:e13077. [PMID: 30721530 DOI: 10.1111/eci.13077] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 01/11/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Smoking remains the most important avoidable cause of global mortality. Even though the number of cigarette smokers declines in first world countries, the uses of alternative nicotine delivery products increase and may even surpass the sells of cigarettes. In this light, the explicit role of nicotine in the development of cardiovascular diseases should be elucidated. OBJECTIVES This narrative review attempts to connect current literature about possible effects of nicotine on the environment of the vasculature to the pathogenesis of vascular calcification, focusing on the tunica media of the vessel wall. METHODS For this review, papers found on Pubmed and Medline until December 2018 by searching for the keywords nicotine, vascular calcification, oxidative stress, osteoblastic transdifferentiation and matrix degradation were considered. RESULTS Nicotine creates an environment that probably facilitates and maybe even induces osteogenic transdifferentiation of VSMC by inflammation, endothelial dysfunction and reactive oxygen species. This process is believed to be a key event in calcification of the tunica media of the vessel wall. Furthermore, nicotine could lead to the formation of nucleation sites for hydroxyapatite by facilitating matrix vesicles and extracellular matrix degradation. CONCLUSIONS There is a growing body of evidence implicating that nicotine alone could impair vascular function and lead to vascular calcification. Further research is necessary to elucidate the explicit influence of nicotine on arteriosclerosis.
Collapse
Affiliation(s)
- Milen Babic
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mirjam Schuchardt
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus Tölle
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus van der Giet
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
33
|
Faggiano P, Dasseni N, Gaibazzi N, Rossi A, Henein M, Pressman G. Cardiac calcification as a marker of subclinical atherosclerosis and predictor of cardiovascular events: A review of the evidence. Eur J Prev Cardiol 2019; 26:1191-1204. [DOI: 10.1177/2047487319830485] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Pompilio Faggiano
- Cardiology Division, Spedali Civili Hospital and University of Brescia, Italy
| | - Nicolò Dasseni
- Cardiology Division, Spedali Civili Hospital and University of Brescia, Italy
| | | | - Andrea Rossi
- Cardiology Division, University of Verona, Italy
| | - Michael Henein
- Department of Public Health and Clinical Medicine, Umea University, Sweden
- St George University, London, UK
- Brunel University, London, UK
| | - Gregg Pressman
- Heart and Vascular Institute, Einstein Medical Center, Philadelphia, USA
| |
Collapse
|
34
|
Wei Y, Wu Y, Zhao R, Zhang K, Midgley AC, Kong D, Li Z, Zhao Q. MSC-derived sEVs enhance patency and inhibit calcification of synthetic vascular grafts by immunomodulation in a rat model of hyperlipidemia. Biomaterials 2019; 204:13-24. [PMID: 30875515 DOI: 10.1016/j.biomaterials.2019.01.049] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/03/2019] [Accepted: 01/20/2019] [Indexed: 02/07/2023]
Abstract
Vascular grafts often exhibit low patency rates in clinical settings due to the pathological environment within the patients requiring the surgery. Mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEVs) have attracted increasing attention. These sEVs contain many potent signaling molecules that play important roles in tissue regeneration, such as microRNA and cytokines. In this study, a sEVs-functionalized vascular graft was developed, and in vivo performance was systematically evaluated in a rat model of hyperlipidemia. Electrospun poly (ε-caprolactone) (PCL) vascular grafts were first modified with heparin, to enhance the anti-thrombogenicity. MSC-derived sEVs were loaded onto the heparinized PCL grafts to obtain functional vascular grafts. As-prepared vascular grafts were implanted to replace a segment of rat abdominal artery (1 cm) for up to 3 months. Results showed that the incorporation of MSC-derived sEVs effectively inhibited thrombosis and calcification, thus enhancing the patency of vascular grafts. Furthermore, regeneration of the endothelium and vascular smooth muscle was markedly enhanced, as attributed to the bioactive molecules within the sEVs, including vascular endothelial growth factor (VEGF), miRNA126, and miRNA145. More importantly, MSC-derived sEVs demonstrated a robust immunomodulatory effect, that is, they induced the transition of macrophages from a pro-inflammatory and atherogenic (M1) phenotype to an anti-inflammatory and anti-osteogenic (M2c) phenotype. This phenotypic switch was confirmed in both in vitro and in vivo analyses. Taken together, these results suggest that fabrication of vascular grafts with immunomodulatory function can provide an effective approach to improve vascular performance and functionality, with translational implication in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Yifan Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Runxia Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Kaiyue Zhang
- Nankai University School of Medicine, Tianjin 300071, PR China
| | - Adam C Midgley
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin 300071, PR China.
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
35
|
Gilham D, Tsujikawa LM, Sarsons CD, Halliday C, Wasiak S, Stotz SC, Jahagirdar R, Sweeney M, Johansson JO, Wong NCW, Kalantar-Zadeh K, Kulikowski E. Apabetalone downregulates factors and pathways associated with vascular calcification. Atherosclerosis 2018; 280:75-84. [PMID: 30476723 DOI: 10.1016/j.atherosclerosis.2018.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/28/2018] [Accepted: 11/07/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS Apabetalone is an inhibitor of bromodomain and extraterminal (BET) proteins. In clinical trials, apabetalone reduced the incidence of major adverse cardiac events (MACE) in patients with cardiovascular disease and reduced circulating factors that promote vascular calcification (VC). Because VC contributes to MACE, effects of apabetalone on pro-calcific processes were examined. METHODS AND RESULTS Apabetalone inhibited extracellular calcium deposition and opposed induction of transdifferentiation markers in human coronary artery vascular smooth muscle cells (VSMCs) under osteogenic culture conditions. Tissue-nonspecific alkaline phosphatase (TNAP) is a key contributor to VC, and apabetalone suppressed osteogenic induction of the mRNA, protein and enzyme activity. The liver is a major source of circulating TNAP, and apabetalone also downregulated TNAP expression in primary human hepatocytes. BRD4, a transcriptional regulator and target of apabetalone, has been linked to calcification. Osteogenic transdifferentiation of VSMCs resulted in disassembly of 100 BRD4-rich enhancers, with concomitant enlargement of remaining enhancers. Apabetalone reduced the size of BRD4-rich enhancers, consistent with disrupting BRD4 association with chromatin. 38 genes were uniquely associated with BRD4-rich enhancers in osteogenic conditions; 11 were previously associated with calcification. Apabetalone reduced levels of BRD4 on many of these enhancers, which correlated with decreased expression of the associated gene. Bioinformatics revealed BRD4 may cooperate with 7 specific transcription factors to promote transdifferentiation and calcification. CONCLUSIONS Apabetalone counters transdifferentiation and calcification of VSMCs via an epigenetic mechanism involving specific transcription factors. The mechanistic findings, combined with evidence from clinical trials, support further development of apabetalone as a therapeutic for VC.
Collapse
|
36
|
He J, Zhong X, Zhao L, Gan H. JAK2/STAT3/BMP-2 axis and NF-κB pathway are involved in erythropoietin-induced calcification in rat vascular smooth muscle cells. Clin Exp Nephrol 2018; 23:501-512. [PMID: 30406500 DOI: 10.1007/s10157-018-1666-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/28/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Vascular calcification is common in chronic kidney disease (CKD) patients, while erythropoietin (EPO) is widely used in the treatment of renal anemia in CKD patients, whether there is a link between the two is still not clear. METHODS The primary rat vascular smooth muscle cells (VSMCs) and CKD rats were treated with EPO and the calcium deposition was observed by alizarin red staining, von Kossa staining and calcium quantification. Activation of JAK2/STAT3/BMP-2 axis and NF-κB signaling pathways was investigated by Western blotting. RESULTS EPO-induced calcium deposition in VSMCs and significantly potentiated calcification in CKD rats. Furthermore, EPO activated JAK2/STAT3/BMP-2 axis, NF-κB pathway and the pro-calcification effect of EPO was partially blocked by the STAT3 inhibitor (Cryptotanshinone) or NF-κB inhibitor (BAY 11-7082), respectively, in vitro. CONCLUSION EPO could promote VSMCs calcification in vitro and in vivo and this effect may be achieved through the JAK2/STAT3/BMP-2 axis and NF-κB pathway.
Collapse
Affiliation(s)
- Jin He
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaoyi Zhong
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lin Zhao
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Hua Gan
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
37
|
Nazemi S, Rezapour A, Moallem SMH, Afshar M, Elyasi S, Mashreghi Moghadam HR, Dargahi Zaboli M, Mohammadpour AH. Could decorin be a biomarker of coronary artery disease? A pilot study in human beings. ACTA BIO-MEDICA : ATENEI PARMENSIS 2018; 89:365-369. [PMID: 30333460 PMCID: PMC6502117 DOI: 10.23750/abm.v89i3.6024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
Background and aim: Nowadays there is a strong necessity in identifying patients who may be exposed to the risk for future cardiovascular events like progressive atherosclerotic disease. Biomarkers are valuable tools for this purpose. Coronary artery calcification (CAC) is utilized as an important tool for the global risk assessment of cardiovascular events in individuals with intermediate risk. Decorin (DCN) is a small leucine-rich proteoglycan that induces calcification of arterial smooth muscle cell and localizes to mineral deposition in human atherosclerotic plaque. The main purpose of this clinical study was to find out the correlation between Decorin serum concentration and CAC in human for the first time. Methods:In this study 84 patients with coronary artery disease who fulfilled inclusion and exclusion criteria, entered the study. For all patients a questionnaire consisting demographic data and traditional cardiovascular risk factors were completed. CT-Angiography was carried out to determine coronary artery calcium score and ELISA method was used for measuring DCN serum concentrations. Results:No significant correlation between DCN serum concentration and total CAC score and also CAC of left anterior descending, right coronary artery, left main coronary artery and circumflex was found in the study population (P>0.05). Conclusions:On the basis of our results DCN serum concentration is not a suitable biomarker of coronary artery disease. However, more studies with higher sample size are necessary for its confirmation. (www.actabiomedica.it)
Collapse
Affiliation(s)
- Saeed Nazemi
- Department of Cardiovascular Diseases, Razavi Hospital, Iran;.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Chattopadhyay A, Yang X, Mukherjee P, Sulaiman D, Fogelman HR, Grijalva V, Dubinett S, Wasler TC, Paul MK, Salehi-Rad R, Mack JJ, Iruela-Arispe ML, Navab M, Fogelman AM, Reddy ST. Treating the Intestine with Oral ApoA-I Mimetic Tg6F Reduces Tumor Burden in Mouse Models of Metastatic Lung Cancer. Sci Rep 2018; 8:9032. [PMID: 29899427 PMCID: PMC5998131 DOI: 10.1038/s41598-018-26755-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/16/2018] [Indexed: 12/16/2022] Open
Abstract
Having demonstrated that apolipoprotein A-I (apoA-I) mimetic peptides ameliorate cancer in mouse models, we sought to determine the mechanism for the anti-tumorigenic function of these peptides. CT-26 cells (colon cancer cells that implant and grow into tumors in the lungs) were injected into wild-type BALB/c mice. The day after injection, mice were either continued on chow or switched to chow containing 0.06% of a concentrate of transgenic tomatoes expressing the apoA-I mimetic peptide 6F (Tg6F). After four weeks, the number of lung tumors was significantly lower in Tg6F-fed mice. Gene expression array analyses of jejunum and lung identified Notch pathway genes significantly upregulated, whereas osteopontin (Spp1) was significantly downregulated by Tg6F in both jejunum and lung. In jejunum, Tg6F increased protein levels for Notch1, Notch2, Dll1, and Dll4. In lung, Tg6F increased protein levels for Notch1 and Dll4 and decreased Spp1. Tg6F reduced oxidized phospholipid levels (E06 immunoreactivity) and reduced 25-hydroxycholesterol (25-OHC) levels, which are known to inhibit Notch1 and induce Spp1, respectively. Notch pathway promotes anti-tumorigenic patrolling monocytes, while Spp1 facilitates pro-tumorigenic myeloid derived suppressor cells (MDSCs) formation. Tg6F-fed mice had higher numbers of patrolling monocytes in jejunum and in lung (p < 0.02), and lower plasma levels of Spp1 with reduced numbers of MDSCs in jejunum and in lung (p < 0.03). We conclude that Tg6F alters levels of specific oxidized lipids and 25-OHC to modulate Notch pathways and Spp1, which alter small intestine immune cells, leading to similar changes in lung that reduce tumor burden.
Collapse
Affiliation(s)
- Arnab Chattopadhyay
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Xinying Yang
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Pallavi Mukherjee
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Dawoud Sulaiman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
- Molecular Toxicology Interdepartmental Degree Program, Fielding School of Public Health, University of California, Los Angeles, CA, 90095-1736, USA
| | - Hannah R Fogelman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Victor Grijalva
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Steven Dubinett
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Tonya C Wasler
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Manash K Paul
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Ramin Salehi-Rad
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Julia J Mack
- Department of Molecular, Cell and Developmental Biology, College of Letters and Science, University of California, Los Angeles, CA, 90095-1736, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology, College of Letters and Science, University of California, Los Angeles, CA, 90095-1736, USA
| | - Mohamad Navab
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA
| | - Srinivasa T Reddy
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA.
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA.
- Molecular Toxicology Interdepartmental Degree Program, Fielding School of Public Health, University of California, Los Angeles, CA, 90095-1736, USA.
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1736, USA.
| |
Collapse
|
39
|
Zhang C, Zhang K, Huang F, Feng W, Chen J, Zhang H, Wang J, Luo P, Huang H. Exosomes, the message transporters in vascular calcification. J Cell Mol Med 2018; 22:4024-4033. [PMID: 29892998 PMCID: PMC6111818 DOI: 10.1111/jcmm.13692] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/18/2018] [Indexed: 12/17/2022] Open
Abstract
Vascular calcification (VC) is caused by hydroxyapatite deposition in the intimal and medial layers of the vascular wall, leading to severe cardiovascular events in patients with hypertension, chronic kidney disease and diabetes mellitus. VC occurrences involve complicated mechanism networks, such as matrix vesicles or exosomes production, osteogenic differentiation, reduced cell viability, aging and so on. However, with present therapeutic methods targeting at VC ineffectively, novel targets for VC treatment are demanded. Exosomes are proven to participate in VC and function as initializers for mineral deposition. Secreted exosomes loaded with microRNAs are also demonstrated to modulate VC procession in recipient vascular smooth muscle cells. In this review, we targeted at the roles of exosomes during VC, especially at their effects on transporting biological information among cells. Moreover, we will discuss the potential mechanisms of exosomes in VC.
Collapse
Affiliation(s)
- Chao Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Kun Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Feifei Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Weijing Feng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Jie Chen
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China.,Department of Radiation Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huanji Zhang
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jingfeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| | - Pei Luo
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Hui Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, GuangZhou, China
| |
Collapse
|
40
|
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
|
41
|
Leszczynska A, Murphy JM. Vascular Calcification: Is it rather a Stem/Progenitor Cells Driven Phenomenon? Front Bioeng Biotechnol 2018; 6:10. [PMID: 29479528 PMCID: PMC5811524 DOI: 10.3389/fbioe.2018.00010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Vascular calcification (VC) has witnessed a surge of interest. Vasculature is virtually an omnipresent organ and has a notably high capacity for repair throughout embryonic and adult life. Of the vascular diseases, atherosclerosis is a leading cause of morbidity and mortality on account of ectopic cartilage and bone formation. Despite the identification of a number of risk factors, all the current theories explaining pathogenesis of VC in atherosclerosis are far from complete. The most widely accepted response to injury theory and smooth muscle transdifferentiation to explain the VC observed in atherosclerosis is being challenged. Recent focus on circulating and resident progenitor cells in the vasculature and their role in atherogenesis and VC has been the driving force behind this review. This review discusses intrinsic cellular players contributing to fate determination of cells and tissues to form ectopic cartilage and bone formation.
Collapse
Affiliation(s)
- Aleksandra Leszczynska
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - J Mary Murphy
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
42
|
Espitia O, Chatelais M, Steenman M, Charrier C, Maurel B, Georges S, Houlgatte R, Verrecchia F, Ory B, Lamoureux F, Heymann D, Gouëffic Y, Quillard T. Implication of molecular vascular smooth muscle cell heterogeneity among arterial beds in arterial calcification. PLoS One 2018; 13:e0191976. [PMID: 29373585 PMCID: PMC5786328 DOI: 10.1371/journal.pone.0191976] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/15/2018] [Indexed: 11/29/2022] Open
Abstract
Vascular calcification is a strong and independent predictive factor for cardiovascular complications and mortality. Our previous work identified important discrepancies in plaque composition and calcification types between carotid and femoral arteries. The objective of this study is to further characterize and understand the heterogeneity in vascular calcification among vascular beds, and to identify molecular mechanisms underlying this process. We established ECLAGEN biocollection that encompasses human atherosclerotic lesions and healthy arteries from different locations (abdominal, thoracic aorta, carotid, femoral, and infrapopliteal arteries) for histological, cell isolation, and transcriptomic analysis. Our results show that lesion composition differs between these locations. Femoral arteries are the most calcified arteries overall. They develop denser calcifications (sheet-like, nodule), and are highly susceptible to osteoid metaplasia. These discrepancies may derive from intrinsic differences between SMCs originating from these locations, as microarray analysis showed specific transcriptomic profiles between primary SMCs isolated from each arterial bed. These molecular differences translated into functional disparities. SMC from femoral arteries showed the highest propensity to mineralize due to an increase in basal TGFβ signaling. Our results suggest that biological heterogeneity of resident vascular cells between arterial beds, reflected by our transcriptomic analysis, is critical in understanding plaque biology and calcification, and may have strong implications in vascular therapeutic approaches.
Collapse
Affiliation(s)
- Olivier Espitia
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
- CHU Hôtel Dieu, Nantes, France
| | - Mathias Chatelais
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
| | - Marja Steenman
- Institut du Thorax, Inserm UMR1087, Faculté de Médecine, Université de Nantes, Nantes Atlantique Universités, Nantes, France
| | - Céline Charrier
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
| | - Blandine Maurel
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
- CHU Hôtel Dieu, Nantes, France
| | - Steven Georges
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
| | - Rémi Houlgatte
- Inserm U954, Faculty of Medicine, Nancy, France, DRCI, University Hospital of Nancy, Nancy, France
| | - Franck Verrecchia
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
| | - Benjamin Ory
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
| | - François Lamoureux
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
| | - Dominique Heymann
- Institut de Cancérologie de l'Ouest, site René Gauducheau, Boulevard Professeur Jacques Monod, Saint-Herblain, France
- University of Sheffield, Department of Oncology and Metabolism, INSERM, European Associated Laboratory “Sarcoma Research Unit”, Medical School, Sheffield, United Kingdom
- University of Nantes, Faculty of Medicine, Nantes, France
| | - Yann Gouëffic
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
- CHU Hôtel Dieu, Nantes, France
| | - Thibaut Quillard
- INSERM, UMR 1238, Nantes, France; Université de Nantes, Nantes Atlantique Universités, Laboratoire « Sarcome osseux et remodelage des tissus osseux calcifiés », Faculté de Médecine, Nantes, France
- CHU Hôtel Dieu, Nantes, France
- * E-mail:
| |
Collapse
|
43
|
Yamauchi Y, Rogers MA. Sterol Metabolism and Transport in Atherosclerosis and Cancer. Front Endocrinol (Lausanne) 2018; 9:509. [PMID: 30283400 PMCID: PMC6157400 DOI: 10.3389/fendo.2018.00509] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/14/2018] [Indexed: 01/22/2023] Open
Abstract
Cholesterol is a vital lipid molecule for mammalian cells, regulating fluidity of biological membranes, and serving as an essential constituent of lipid rafts. Mammalian cells acquire cholesterol from extracellular lipoproteins and from de novo synthesis. Cholesterol biosynthesis generates various precursor sterols. Cholesterol undergoes metabolic conversion into oxygenated sterols (oxysterols), bile acids, and steroid hormones. Cholesterol intermediates and metabolites have diverse and important cellular functions. A network of molecular machineries including transcription factors, protein modifiers, sterol transporters/carriers, and sterol sensors regulate sterol homeostasis in mammalian cells and tissues. Dysfunction in metabolism and transport of cholesterol, sterol intermediates, and oxysterols occurs in various pathophysiological settings such as atherosclerosis, cancers, and neurodegenerative diseases. Here we review the cholesterol, intermediate sterol, and oxysterol regulatory mechanisms and intracellular transport machineries, and discuss the roles of sterols and sterol metabolism in human diseases.
Collapse
Affiliation(s)
- Yoshio Yamauchi
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- *Correspondence: Yoshio Yamauchi
| | - Maximillian A. Rogers
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
44
|
Borland SJ, Morris TG, Borland SC, Morgan MR, Francis SE, Merry CL, Canfield AE. Regulation of vascular smooth muscle cell calcification by syndecan-4/FGF-2/PKCα signalling and cross-talk with TGFβ. Cardiovasc Res 2017; 113:1639-1652. [PMID: 29016732 PMCID: PMC5852548 DOI: 10.1093/cvr/cvx178] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 02/01/2017] [Accepted: 09/04/2017] [Indexed: 11/12/2022] Open
Abstract
AIMS Vascular calcification is a major cause of morbidity and mortality. Fibroblast growth factor-2 (FGF-2) plays an instructive role in osteogenesis and bone development, but its role in vascular calcification was unknown. Therefore, we investigated the involvement of FGF-2 in vascular calcification and determined the mechanism by which it regulates this process. METHODS AND RESULTS We demonstrate that FGF-2 expression is increased in vascular smooth muscle cells (VSMCs) induced to deposit a mineralized matrix by incubation with β-glycerophosphate. FGF-2 is also localized to sites of calcification within human atherosclerotic plaques. The expression of syndecan-4, a heparan sulfate proteoglycan which regulates FGF-2 signalling, is also increased in mineralizing VSMCs and co-localizes with FGF-2 in human calcified atherosclerotic plaques. Exogenous FGF-2 inhibits VSMC mineralization, and this inhibition is reduced when syndecan-4 expression is knocked-down using siRNA. Biochemical inhibition of FGFR signalling using a pan FGFR inhibitor (BGJ398) or knocking-down syndecan-4 expression in VSMCs using siRNA increases VSMC mineralization. These increases are prevented by inhibiting transforming growth factor-β (TGFβ) signalling with SB431542, suggesting cross-talk between FGF-2 and TGFβ signalling is crucial for the regulation of VSMC mineralization. Syndecan-4 can also regulate FGF-2 signalling directly via protein kinase Cα (PKCα) activation. Biochemical inhibition of PKCα activity using Gö6976, or siRNA-mediated suppression of PKCα expression increases VSMC mineralization; this increase is also prevented with SB431542. Finally, the ability of FGF-2 to inhibit VSMC mineralization is reduced when PKCα expression is knocked-down. CONCLUSION This is the first demonstration that syndecan-4 promotes FGF-2 signalling, and in turn, suppresses VSMC mineralization by down-regulating TGFβ signalling. Our discoveries that FGF-2 and syndecan-4 expression is increased in mineralizing VSMCs and that PKCα regulates FGF-2 and TGFβ signalling in VSMCs suggests that the syndecan-4/FGF-2/TGFβ signalling axis could represent a new therapeutic target for vascular calcification.
Collapse
Affiliation(s)
- Samantha J. Borland
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Stem Cell Glycobiology Group, School of Materials, University of Manchester, Manchester, UK
| | - Thomas G. Morris
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Shona C. Borland
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Mark R. Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Sheila E. Francis
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Catherine L.R. Merry
- Stem Cell Glycobiology Group, School of Materials, University of Manchester, Manchester, UK
- Wolfson Centre for Stem Cells, Tissue Engineering & Modelling, Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Ann E. Canfield
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
45
|
Current knowledge on the mechanism of atherosclerosis and pro-atherosclerotic properties of oxysterols. Lipids Health Dis 2017; 16:188. [PMID: 28969682 PMCID: PMC5625595 DOI: 10.1186/s12944-017-0579-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/22/2017] [Indexed: 01/22/2023] Open
Abstract
Due to the fact that one of the main causes of worldwide deaths are directly related to atherosclerosis, scientists are constantly looking for atherosclerotic factors, in an attempt to reduce prevalence of this disease. The most important known pro-atherosclerotic factors include: elevated levels of LDL, low HDL levels, obesity and overweight, diabetes, family history of coronary heart disease and cigarette smoking. Since finding oxidized forms of cholesterol – oxysterols – in lesion in the arteries, it has also been presumed they possess pro-atherosclerotic properties. The formation of oxysterols in the atherosclerosis lesions, as a result of LDL oxidation due to the inflammatory response of cells to mechanical stress, is confirmed. However, it is still unknown, what exactly oxysterols cause in connection with atherosclerosis, after gaining entry to the human body e.g., with food containing high amounts of cholesterol, after being heated. The in vivo studies should provide data to finally prove or disprove the thesis regarding the pro-atherosclerotic prosperities of oxysterols, yet despite dozens of available in vivo research some studies confirm such properties, other disprove them. In this article we present the current knowledge about the mechanism of formation of atherosclerotic lesions and we summarize available data on in vivo studies, which investigated whether oxysterols have properties to cause the formation and accelerate the progress of the disease. Additionally we will try to discuss why such different results were obtained in all in vivo studies.
Collapse
|
46
|
Arzani A, Masters KS, Mofrad MRK. Multiscale Systems Biology Model of Calcific Aortic Valve Disease Progression. ACS Biomater Sci Eng 2017; 3:2922-2933. [DOI: 10.1021/acsbiomaterials.7b00174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Amirhossein Arzani
- Molecular
Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical
Engineering, University of California, Berkeley, California 94720, United States
| | - Kristyn S. Masters
- Department
of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Mohammad R. K. Mofrad
- Molecular
Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical
Engineering, University of California, Berkeley, California 94720, United States
| |
Collapse
|
47
|
Oestrogen Inhibits Arterial Calcification by Promoting Autophagy. Sci Rep 2017; 7:3549. [PMID: 28615727 PMCID: PMC5471178 DOI: 10.1038/s41598-017-03801-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 05/04/2017] [Indexed: 01/08/2023] Open
Abstract
Arterial calcification is a major complication of cardiovascular disease. Oestrogen replacement therapy in postmenopausal women is associated with lower levels of coronary artery calcification, but its mechanism of action remains unclear. Here, we show that oestrogen inhibits the osteoblastic differentiation of vascular smooth muscle cells (VSMCs) in vitro and arterial calcification in vivo by promoting autophagy. Through electron microscopy, GFP–LC3 redistribution, and immunofluorescence analyses as well as measurement of the expression of the autophagosome marker light-chain I/II (LC3I/II) and autophagy protein 5 (Atg5), we show that autophagy is increased in VSMCs by oestrogen in vitro and in vivo. The inhibitory effect of oestrogen on arterial calcification was counteracted by 3-methyladenine (3MA) or knockdown of Atg5 and was increased by rapamycin. Furthermore, the inhibitory effect of oestrogen on arterial calcification and the degree of autophagy induced by oestrogen were blocked by a nonselective oestrogen receptor (ER) antagonist (ICI 182780), a selective oestrogen receptor alpha (ERα) antagonist (MPP), and ERα-specific siRNA. Our data indicate that oestrogen inhibits the osteoblastic differentiation of VSMCs by promoting autophagy through the ERα signalling pathway in vitro and arterial calcification in vivo by increasing autophagy. Our findings provide new insights into the mechanism by which oestrogen contributes to vascular calcification in vitro and in vivo.
Collapse
|
48
|
Lau I, Potluri A, Ibeh CL, Redman RS, Paal E, Bandyopadhyay BC. Microcalcifications in stone-obstructed human submandibular gland are associated with apoptosis and cell proliferation. Arch Oral Biol 2017. [PMID: 28623687 DOI: 10.1016/j.archoralbio.2017.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Human submandibular gland (SMG) stones are associated with inflammation, fibrosis and microcalcifications in the surrounding tissues. However, there is little information about the accompanying cell injury-repair process, apoptosis, and cell proliferation. The purpose of this study was to investigate such an association and its clinical significance. DESIGN OF STUDY Mid-gland paraffin sections of human SMGs ("stone glands") and normal SMGs ("non-stone glands") were subjected to stains for general histology (hematoxylin and eosin), fibrosis (Masson's trichrome), and calcification (alizarin red) and to immunohistochemistry for proliferative activity (Ki-67), and apoptosis (Caspase-3). Tissues were assessed for areas of inflammation, calcium deposition, and fibrosis, and for cycling and apoptotic cells. RESULTS Acini were atrophic and proportionately fewer in lobules with fibrosis in stone glands. Additionally, stone glands had intraluminal calcifications (microliths) in scattered excretory and striated ducts and blood vessel walls. Areas of inflammation and fibrosis were small and uncommon, and calcifications were not seen in non-stone glands. Proliferating and apoptotic cells were common in the main duct of stone glands where ciliated and mucous cell hyperplasia and stratified squamous metaplasia had occurred, uncommon in the main duct of non-stone glands, and uncommon in all other parenchymal elements of both stone and non-stone glands. CONCLUSION Stone obstruction in the main excretory ducts of SMG resulted in progressive depletion of acini from proximal to distal lobules via calcification, inflammation, fibrosis, and parenchymal cell atrophy, apoptosis and proliferation. Interlobular duct microliths contributed to this depletion by further provoking intralobular inflammation, fibrosis, and acinar atrophy.
Collapse
Affiliation(s)
- Ivan Lau
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, Washington DC, United States
| | - Ajay Potluri
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, Washington DC, United States
| | - Cliff-Lawrence Ibeh
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, Washington DC, United States
| | - Robert S Redman
- Oral Pathology Research Laboratory, Research Service, Veterans Affairs Medical Center, Washington DC, United States
| | - Edina Paal
- Pathology and Laboratory Service, Veterans Affairs Medical Center, Washington DC, United States
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, Washington DC, United States; Department of Pharmacology and Physiology, Georgetown University, Washington DC, United States.
| |
Collapse
|
49
|
Song R, Fullerton DA, Ao L, Zhao KS, Meng X. An epigenetic regulatory loop controls pro-osteogenic activation by TGF-β1 or bone morphogenetic protein 2 in human aortic valve interstitial cells. J Biol Chem 2017; 292:8657-8666. [PMID: 28377507 DOI: 10.1074/jbc.m117.783308] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/03/2017] [Indexed: 01/04/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is common in the elderly population, but pharmacological interventions for managing valvular calcification are unavailable. Transforming growth factor β1 (TGF-β1) and bone morphogenetic protein 2 (BMP-2) induce pro-osteogenic activation of human aortic valve interstitial cells (AVICs) that play an important role in valvular calcification. However, the molecular mechanism underlying pro-osteogenic activation in AVICs is incompletely understood. Here, we investigated an epigenetic regulatory mechanism in human AVIC pro-osteogenic activation induced by TGF-β1 and BMP-2. Microarray and real-time PCR analyses revealed that microRNA (miR)-486 up-regulation and miR-204 down-regulation were characteristic changes in TGF-β1- and BMP-2-stimulated normal AVICs and in AVICs from calcified valves. Both TGF-β1 and BMP-2 down-regulated miR-204 through Smad pathways. Interestingly, an miR-486 antagomir diminished the effect of TGF-β1 and BMP-2 on miR-204 levels and calcium deposit formation. Furthermore, the miR-486 antagomir increased the expression of Smurf2, a Smad inhibitor, in the presence or absence of TGF-β1 or BMP-2 stimulation, whereas a miR-486 mimic reduced Smurf2 expression. Smurf2 knockdown augmented TGF-β1- or BMP-2-induced miR-204 down-regulation and resulted in increased expression of the osteoblastic biomarkers Osx and Runx2. In summary, we found that TGF-β1 and BMP-2 up-regulate miR-486 and down-regulate miR-204 in human AVICs to promote pro-osteogenic activity and that miR-486 inhibits Smurf2 expression to augment the miR-204 down-regulation. We conclude that the miR-486-Smurf2-Smad loop plays an important role in regulating AVIC pro-osteogenic activation in response to TGF-β1 or BMP-2. Targeting this regulatory loop may have therapeutic potential for suppressing aortic valve calcification.
Collapse
Affiliation(s)
- Rui Song
- From the Department of Surgery, University of Colorado Denver, Aurora, Colorado 80045 and
| | - David A Fullerton
- From the Department of Surgery, University of Colorado Denver, Aurora, Colorado 80045 and
| | - Lihua Ao
- From the Department of Surgery, University of Colorado Denver, Aurora, Colorado 80045 and
| | - Ke-Seng Zhao
- the Department of Pathophysiology, Guangdong Key Laboratory of Shock and Microcirculation Research, Southern Medical University, Guangzhou 510515, China
| | - Xianzhong Meng
- From the Department of Surgery, University of Colorado Denver, Aurora, Colorado 80045 and
| |
Collapse
|
50
|
Tanaka T, Hatakeyama S, Yamamoto H, Narita T, Hamano I, Matsumoto T, Soma O, Tobisawa Y, Yoneyama T, Yoneyama T, Hashimoto Y, Koie T, Takahashi I, Nakaji S, Terayama Y, Funyu T, Ohyama C. Clinical relevance of aortic calcification in urolithiasis patients. BMC Urol 2017; 17:25. [PMID: 28376750 PMCID: PMC5379761 DOI: 10.1186/s12894-017-0218-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 03/27/2017] [Indexed: 12/31/2022] Open
Abstract
Background The aim of the present study is to investigate the clinical relevance of aortic calcification in urolithiasis patients. Methods Between January 2010 and September 2014, 1221 patients with urolithiasis were treated in Oyokyo Kidney Research Institute and Hirosaki University Hospital. Among these, 287 patients (Stone group) on whom adequate data were available were included in this retrospective study. We also selected 148 subjects with early stage (pT1N0M0) renal cell carcinoma from 607 renal cell carcinoma patients who underwent radical nephrectomy at Hirosaki University Hospital (Non-stone group) as control subjects. Validity of the Non-stone group was evaluated by comparison with pair-matched 296 volunteers from 1166 subjects who participated in the Iwaki Health Promotion Project in 2014. Thereafter, age, body mass index, aortic calcification index (ACI), renal function, serum uric acid concentrations, and comorbidities (diabetes, hypertension, or cardiovascular disease) were compared between the Non-stone and Stone groups. Independent factors for higher ACI and impaired renal function were assessed using multivariate logistic regression analysis. Results We confirmed relevance of Non-stone group patients as a control subject by comparing the pair-matched community-dwelling volunteers. Backgrounds of patients between the Non-stone and Stone groups were not significantly different except for the presence of hypertension in the Stone group. ACI was not significantly high in the Stone group compared with the Non-stone group. However, age-adjusted ACI was greater in the Stone group than the Non-stone group. Among urolithiasis patients, ACI was significantly higher in uric acid containing stone patients. The number of patients with stage 3B chronic kidney disease (CKD) was significantly higher in the Stone group than in the Non-stone group (12% vs. 4%, P = 0.008). Multivariate logistic regression analysis showed higher aortic calcification index (>13%), and being a stone former were independent factors for stage 3B CKD at the time of diagnosis. Conclusion Aortic calcification and being a stone former had harmful influence on renal function. This study was registered as a clinical trial: UMIN: UMIN000022962.
Collapse
Affiliation(s)
- Toshikazu Tanaka
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Shingo Hatakeyama
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan.
| | - Hayato Yamamoto
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Takuma Narita
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Itsuto Hamano
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Teppei Matsumoto
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Osamu Soma
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Yuki Tobisawa
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Tohru Yoneyama
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Takahiro Yoneyama
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Yasuhiro Hashimoto
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Takuya Koie
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan
| | - Ippei Takahashi
- Department of Social Medicine, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Shigeyuki Nakaji
- Department of Social Medicine, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Yuriko Terayama
- Department of Urology, Oyokyo Kidney Research Institute, Hirosaki, Japan
| | - Tomihisa Funyu
- Department of Urology, Oyokyo Kidney Research Institute, Hirosaki, Japan
| | - Chikara Ohyama
- Department of Urology, Hirosaki University Graduate School of Medicine, 5 Zaifu-chou, Hirosaki, 036-8562, Japan.,Department of Advanced Transplant and Regenerative Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|