1
|
Wu M, Li H, Sun X, Zhong R, Cai L, Chen R, Madeniyet M, Ren K, Peng Z, Yang Y, Chen W, Tu Y, Lai M, Deng J, Wu Y, Zhao S, Ruan Q, Rao M, Xie S, Ye Y, Wan J. Aerobic exercise prevents renal osteodystrophy via irisin-activated osteoblasts. JCI Insight 2025; 10:e184468. [PMID: 39883525 PMCID: PMC11949034 DOI: 10.1172/jci.insight.184468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Renal osteodystrophy is commonly seen in patients with chronic kidney disease (CKD) due to disrupted mineral homeostasis. Given the impaired renal function in these patients, common antiresorptive agents, including bisphosphonates, must be used with caution or even contraindicated. Therefore, an alternative therapy without renal burden to combat renal osteodystrophy is urgently needed. Here, we report that clinically relevant aerobic exercise significantly prevents high-turnover renal osteodystrophy in CKD mice and patients with CKD without compromising renal function. Mechanistically, 4-week aerobic exercise in CKD mice increased expression of skeletal muscle PPARγ coactivator-1α (PGC-1α) and circulating irisin. Both exercise and irisin administration significantly activated osteoblasts, but not osteoclasts, via integrin αvβ5, thereby conferring bone quality benefits. Removal of irisin-influenced thermogenic adipose tissues or genetic ablation of uncoupling protein 1 did not alter the irisin-conferred antiosteodystrophy effect. Importantly, in a pilot clinical study, 12-week aerobic exercise in patients with high-grade CKD significantly increased circulating irisin and prevented osteodystrophy progression, without detectable renal burden. The combination of irisin and current antiresorptive agents effectively rescued renal osteodystrophy in mice. Our work provides mechanistic insights into the role of exercise and irisin in renal osteodystrophy, and it highlights a clinically relevant, low-cost, kidney-friendly therapy for patients with this devastating disease.
Collapse
Affiliation(s)
- Meng Wu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Huilan Li
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoting Sun
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Rongrong Zhong
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Linli Cai
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ruibo Chen
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Madiya Madeniyet
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kana Ren
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhen Peng
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yujie Yang
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Weiqin Chen
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yanling Tu
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Miaoxin Lai
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Jinxiu Deng
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yuting Wu
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Shumin Zhao
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Qingyan Ruan
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Mei Rao
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Sisi Xie
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ying Ye
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, and
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
2
|
Xu L, Liu B, Ma H, Qi E, Ma J, Chang T, Zhang J, Zhang W, Chen W, Cao X, Xiong X. O-GlcNAc transferase promotes vascular smooth muscle calcification through modulating Wnt/β-catenin signaling. FASEB J 2024; 38:e70271. [PMID: 39704274 DOI: 10.1096/fj.202401649rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Vascular calcification (VC), associated with high cardiovascular mortality in patients with chronic kidney disease (CKD), involves osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs). O-GlcNAcylation, a dynamic post-translational modification, is closely linked to cardiovascular diseases, including VC. However, the exact role and molecular mechanism of O-GlcNAc signaling in abnormal mineral metabolism-induced VC remain unclear. In the current study, we found that the levels of O-GlcNAc transferase (OGT) and global protein O-GlcNAcylation were significantly upregulated in the artery tissues of mouse calcification models and CKD patients with VC. To further delineate the in vivo role of OGT in VC, we generated Ogt smooth muscle cell-specific knockout mice and challenged them with 5/6 nephrectomy (5/6 Nx) or high-dose vitamin D3 to induce VC. Deletion of Ogt in VSMCs led to alleviated VC in response to 5/6 Nx or VD3. Moreover, elevated O-GlcNAcylation, induced by Thiamet-G, facilitated osteogenic transdifferentiation in VSMCs in response to phosphate, whereas OSMI-1, which reduces O-GlcNAcylation, exhibited an opposite phenotypic effect. Mechanistically, O-GlcNAc signaling enhanced the osteogenic conversion of VSMCs through regulation of canonical Wnt/β-catenin pathway. Indeed, β-catenin was O-GlcNAcylated by OGT and further increased its transcriptional activity in VSMCs. Furthermore, pharmacological activation of Wnt/β-catenin signaling largely reversed the diminished aortic calcification caused by Ogt ablation. Our findings demonstrate that smooth muscle O-GlcNAc signaling plays an important role in regulating hyperphosphatemia-induced VC and reveal that O-GlcNAcylation of β-catenin protein modulates its content and activity in VSMCs.
Collapse
MESH Headings
- Animals
- N-Acetylglucosaminyltransferases/metabolism
- N-Acetylglucosaminyltransferases/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Wnt Signaling Pathway
- Mice
- Mice, Knockout
- Humans
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Male
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- beta Catenin/metabolism
- Cells, Cultured
- Osteogenesis
Collapse
Affiliation(s)
- Lin Xu
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Boao Liu
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Honghui Ma
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Enbo Qi
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jie Ma
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Tingmin Chang
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinghong Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wencheng Zhang
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Weiqian Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xuan Cao
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Xiwen Xiong
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
3
|
Wang X, Ren J, Fang F, Wang E, Li J, He W, Zhang Z, Shen Y, Liu X. Matrix vesicles from osteoblasts promote atherosclerotic calcification. Matrix Biol 2024; 134:79-92. [PMID: 39580186 DOI: 10.1016/j.matbio.2024.09.003] [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: 06/05/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 11/25/2024]
Abstract
Atherosclerotic calcification often coincides with osteoporosis, suggesting a potential interplay between bone and vascular mineralization. Osteoblast-derived matrix vesicles (Ost-MVs), pivotal in bone mineralization, have emerged as potential contributors to ectopic vascular calcification. However, the precise role of Ost-MVs in vascular calcification and the underlying mechanisms remain elusive. In this study, we observed a concomitant increase in atherosclerotic calcification and bone loss, accompanied by elevated release of Ost-MVs into circulation. We demonstrate that circulating Ost-MVs target plaque lesions in the setting of atherosclerosis. Mechanistically, vascular injury facilitates transendothelial transport of Ost-MVs, collagen І remodeling promotes Ost-MVs aggregation, and vascular smooth muscle cell (VSMC) phenotypic switching enhances MV uptake. These pathological changes during atherosclerosis collectively contribute to Ost-MVs recruitment into the vasculature. Furthermore, Ost-MVs and VSMC-derived matrix vesicles (VSMC-MVs) exacerbate calcification via the Ras-Raf-ERK pathway. Our findings unveil a novel Ost-MVs-mediated mechanism participating in vascular calcification and enriching our understanding of bone-vascular crosstalk.
Collapse
Affiliation(s)
- Xiaoli Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Jie Ren
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Erxiang Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Jianwei Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, PR China
| | - Weihong He
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, PR China.
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China.
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China.
| |
Collapse
|
4
|
Flores-Roco A, Lago BM, Villa-Bellosta R. Elevated glucose levels increase vascular calcification risk by disrupting extracellular pyrophosphate metabolism. Cardiovasc Diabetol 2024; 23:405. [PMID: 39529124 PMCID: PMC11555999 DOI: 10.1186/s12933-024-02502-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Vascular calcification is a major contributor to cardiovascular disease, especially diabetes, where it exacerbates morbidity and mortality. Although pyrophosphate is a recognized natural inhibitor of vascular calcification, there have been no prior studies examining its specific deficiency in diabetic conditions. This study is the first to analyze the direct link between elevated glucose levels and disruptions in extracellular pyrophosphate metabolism. METHODS Rat aortic smooth muscle cells, streptozotocin (STZ)-induced diabetic rats, and diabetic human aortic smooth muscle cells were used to assess the effects of elevated glucose levels on pyrophosphate metabolism and vascular calcification. The techniques used include extracellular pyrophosphate metabolism assays, thin-layer chromatography, phosphate-induced calcification assays, BrdU incorporation for DNA synthesis, aortic smooth muscle cell viability and proliferation assays, and quantitative PCR for enzyme expression analysis. Additionally, extracellular pyrophosphate metabolism was examined through the use of radiolabeled isotopes to track ATP and pyrophosphate transformations. RESULTS Elevated glucose led to a significant reduction in extracellular pyrophosphate across all diabetic models. This metabolic disruption was marked by notable downregulation of both the expression and activity of ectonucleotide pyrophosphatase/phosphodiesterase 1, a key enzyme that converts ATP to pyrophosphate. We also observed an upregulation of ectonucleoside triphosphate diphosphohydrolase 1, which preferentially hydrolyzes ATP to inorganic phosphate rather than pyrophosphate. Moreover, tissue-nonspecific alkaline phosphatase activity was markedly elevated across all diabetic models. This shift in enzyme activity significantly reduced the pyrophosphate/phosphate ratio. In addition, we noted a marked downregulation of matrix Gla protein, another inhibitor of vascular calcification. The impaired pyrophosphate metabolism was further corroborated by calcification experiments across all three diabetic models, which demonstrated an increased propensity for vascular calcification. CONCLUSIONS This study demonstrated that diabetes-induced high glucose disrupts extracellular pyrophosphate metabolism, compromising its protective role against vascular calcification. These findings identify pyrophosphate deficiency as a potential mechanism in diabetic vascular calcification, highlighting a new therapeutic target. Strategies aimed at restoring or enhancing pyrophosphate levels may offer significant potential in mitigating cardiovascular complications in diabetic patients, meriting further investigation.
Collapse
MESH Headings
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Diphosphates/metabolism
- Animals
- Humans
- Phosphoric Diester Hydrolases/metabolism
- Diabetes Mellitus, Experimental/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Pyrophosphatases/metabolism
- Pyrophosphatases/genetics
- Cells, Cultured
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Rats, Sprague-Dawley
- Alkaline Phosphatase/metabolism
- Alkaline Phosphatase/blood
- Extracellular Matrix Proteins/metabolism
- Extracellular Matrix Proteins/genetics
- Blood Glucose/metabolism
- Matrix Gla Protein
- Calcium-Binding Proteins/metabolism
- Calcium-Binding Proteins/genetics
- Cell Proliferation/drug effects
- GPI-Linked Proteins/metabolism
- Rats
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/prevention & control
- Aorta/metabolism
- Aorta/pathology
- Aorta/drug effects
- 5'-Nucleotidase
Collapse
Affiliation(s)
- Alicia Flores-Roco
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Campus Vida, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Travesia da Choupana S/N, 15706, Santiago de Compostela, Spain
| | - Belinda M Lago
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Campus Vida, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Travesia da Choupana S/N, 15706, Santiago de Compostela, Spain
| | - Ricardo Villa-Bellosta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Campus Vida, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Health Research Institute of Santiago de Compostela (IDIS), Travesia da Choupana S/N, 15706, Santiago de Compostela, Spain.
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Campus Vida, University of Santiago de Compostela, Avenida de Barcelona S/N, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
5
|
Liang X, Li Y, Wang P, Liu H. Key regulators of vascular calcification in chronic kidney disease: Hyperphosphatemia, BMP2, and RUNX2. PeerJ 2024; 12:e18063. [PMID: 39308809 PMCID: PMC11416758 DOI: 10.7717/peerj.18063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Vascular calcification is quite common in patients with end-stage chronic kidney disease and is a major trigger for cardiovascular complications in these patients. These complications significantly impact the survival rate and long-term prognosis of individuals with chronic kidney disease. Numerous studies have demonstrated that the development of vascular calcification involves various pathophysiological mechanisms, with the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) being of utmost importance. High phosphate levels, bone morphogenetic protein 2 (BMP2), and runt-related transcription factor 2 (RUNX2) play crucial roles in the osteogenic transdifferentiation process of VSMCs. This article primarily reviews the molecular mechanisms by which high phosphate, BMP2, and RUNX2 regulate vascular calcification secondary to chronic kidney disease, and discusses the complex interactions among these factors and their impact on the progression of vascular calcification. The insights provided here aim to offer new perspectives for future research on the phenotypic switching and osteogenic transdifferentiation of VSMCs, as well as to aid in optimizing clinical treatment strategies for this condition, bearing significant clinical and scientific implications.
Collapse
Affiliation(s)
- Xinhua Liang
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Zhanjiang, Guangdong Province, China
| | - Yankun Li
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Zhanjiang, Guangdong Province, China
| | - Peng Wang
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, Guangdong, China
| | - Huafeng Liu
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, Guangdong, China
| |
Collapse
|
6
|
Dhulkifle H, Diab MI, Algonaiah M, Korashy HM, Maayah ZH. Apabetalone (RVX-208): A Potential Epigenetic Therapy for the Treatment of Cardiovascular, Renal, Neurological, Viral, and Cancer Disorders. ACS Pharmacol Transl Sci 2024; 7:546-559. [PMID: 38481679 PMCID: PMC10928887 DOI: 10.1021/acsptsci.3c00219] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/25/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2025]
Abstract
Bromodomain and extra-terminal domain proteins (BET proteins) are epigenetic reader proteins that have been implicated in regulating gene expression through binding to chromatin and interaction with transcription factors. These proteins are located in the nucleus and are responsible for recognizing acetylated lysine residues on histones, reading epigenetic messages, recruiting key transcription factors, and thereby regulating gene expression. BET proteins control the transcription of genes responsible for maladaptive effects in inflammation, cancer, and renal and cardiovascular diseases. Given the multifaceted role of BET proteins in the pathogenesis of various diseases, several small molecule inhibitors of BET proteins have been developed as potential therapeutic targets for treating different diseases in recent years. However, while many nonselective BET inhibitors are indicated for the treatment of cancer, a selective BET inhibitor, apabetalone, is the only oral BET inhibitor in phase III clinical trials for the treatment of cardiovascular diseases and others. Thus, this review aims to present and discuss the preclinical and clinical evidence for the beneficial effects and mechanism of action of apabetalone for treating various diseases.
Collapse
Affiliation(s)
- Hevna Dhulkifle
- Department
of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Mohammad Issam Diab
- Department
of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Majed Algonaiah
- Department
of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Hesham M. Korashy
- Department
of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Zaid H. Maayah
- Department
of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| |
Collapse
|
7
|
Abreu MAD, de Castro PASV, Moreira FRC, de Oliveira Ferreira H, Simões E Silva AC. Potential Role of Novel Cardiovascular Biomarkers in Pediatric Patients with Chronic Kidney Disease. Mini Rev Med Chem 2024; 24:491-506. [PMID: 37231748 DOI: 10.2174/1389557523666230523114331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/15/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Cardiovascular Disease is the leading cause of death in adult and pediatric patients with Chronic Kidney Disease (CKD) and its pathogenesis involves the interaction of multiple pathways. As Inflammatory mechanisms play a critical role in the vascular disease of CKD pediatric patients, there are several biomarkers related to inflammation strongly associated with this comorbidity. OBJECTIVE This review provides available evidence on the link between several biomarkers and the pathophysiology of heart disease in patients with CKD. METHODS The data were obtained independently by the authors, who carried out a comprehensive and non-systematic search in PubMed, Cochrane, Scopus, and SciELO databases. The search terms were "Chronic Kidney Disease", "Cardiovascular Disease", "Pediatrics", "Pathophysiology", "Mineral and Bone Disorder (MBD)", "Renin Angiotensin System (RAS)", "Biomarkers", "BNP", "NTproBNP", "CK-MB", "CXCL6", "CXCL16", "Endocan-1 (ESM-1)", "FABP3", "FABP4", h-FABP", "Oncostatin- M (OSM)", "Placental Growth Factor (PlGF)" and "Troponin I". RESULTS The pathogenesis of CKD-mediated cardiovascular disease is linked to inflammatory biomarkers, which play a critical role in the initiation, maintenance, and progression of cardiovascular disease. There are several biomarkers associated with cardiovascular disease in pediatric patients, including BNP, NTproBNP, CK-MB, CXCL6, CXCL16, Endocan-1 (ESM-1), FABP3, FABP4, Oncostatin- M (OSM), Placental Growth Factor (PlGF), and Troponin I. CONCLUSION The pathogenesis of CKD-mediated cardiovascular disease is not completely understood, but it is linked to inflammatory biomarkers. Further studies are required to elucidate the pathophysiological and potential role of these novel biomarkers.
Collapse
Affiliation(s)
- Maria Augusta Duarte Abreu
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Alves Soares Vaz de Castro
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Rocha Chaves Moreira
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Henrique de Oliveira Ferreira
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Department of Pediatric Unit of Pediatric Nephrology, Faculty of Medicine UFMG, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
8
|
Mebarek S, Buchet R, Pikula S, Strzelecka-Kiliszek A, Brizuela L, Corti G, Collacchi F, Anghieri G, Magrini A, Ciancaglini P, Millan JL, Davies O, Bottini M. Do Media Extracellular Vesicles and Extracellular Vesicles Bound to the Extracellular Matrix Represent Distinct Types of Vesicles? Biomolecules 2023; 14:42. [PMID: 38254642 PMCID: PMC10813234 DOI: 10.3390/biom14010042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Mineralization-competent cells, including hypertrophic chondrocytes, mature osteoblasts, and osteogenic-differentiated smooth muscle cells secrete media extracellular vesicles (media vesicles) and extracellular vesicles bound to the extracellular matrix (matrix vesicles). Media vesicles are purified directly from the extracellular medium. On the other hand, matrix vesicles are purified after discarding the extracellular medium and subjecting the cells embedded in the extracellular matrix or bone or cartilage tissues to an enzymatic treatment. Several pieces of experimental evidence indicated that matrix vesicles and media vesicles isolated from the same types of mineralizing cells have distinct lipid and protein composition as well as functions. These findings support the view that matrix vesicles and media vesicles released by mineralizing cells have different functions in mineralized tissues due to their location, which is anchored to the extracellular matrix versus free-floating.
Collapse
Affiliation(s)
- Saida Mebarek
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR CNRS 5246, Université de Lyon, Université Claude Bernard Lyon 1, 69 622 Villeurbanne Cedex, France; (R.B.); (L.B.)
| | - Rene Buchet
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR CNRS 5246, Université de Lyon, Université Claude Bernard Lyon 1, 69 622 Villeurbanne Cedex, France; (R.B.); (L.B.)
| | - Slawomir Pikula
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (S.P.); (A.S.-K.)
| | - Agnieszka Strzelecka-Kiliszek
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (S.P.); (A.S.-K.)
| | - Leyre Brizuela
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR CNRS 5246, Université de Lyon, Université Claude Bernard Lyon 1, 69 622 Villeurbanne Cedex, France; (R.B.); (L.B.)
| | - Giada Corti
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (G.C.); (F.C.)
| | - Federica Collacchi
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (G.C.); (F.C.)
| | - Genevieve Anghieri
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE113TU, UK; (G.A.); (O.D.)
| | - Andrea Magrini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil;
| | - Jose Luis Millan
- Sanford Children’s Health Research Center, Sanford Burnham Prebys, La Jolla, CA 92037, USA;
| | - Owen Davies
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE113TU, UK; (G.A.); (O.D.)
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (G.C.); (F.C.)
- Sanford Children’s Health Research Center, Sanford Burnham Prebys, La Jolla, CA 92037, USA;
| |
Collapse
|
9
|
Wu X, Shen S, Wu J, Wu S, Wang S, Di F. ENPP1 ameliorates vascular calcification via inhibiting the osteogenic transformation of VSMCs and generating PPi. Open Med (Wars) 2023; 18:20230861. [PMID: 38152331 PMCID: PMC10751896 DOI: 10.1515/med-2023-0861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 12/29/2023] Open
Abstract
This study aims to investigate the impact of ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) on vascular calcification in rats. The rationale behind studying ENPP1's role in vascular calcification lies in its potential to modulate calcification processes. Understanding this relationship can offer insights into novel therapeutic avenues for addressing vascular calcification-related disorders. In this experiment, vascular smooth muscle cell (VSMC) calcification was induced using β-glycerophosphoric acid. Subsequently, recombinant AAV9-carrying ENPP1 was introduced into VSMCs to achieve both in vitro and in vivo overexpression of ENPP1. The findings indicate that ENPP1 overexpression significantly reduces calcium and phosphorus content in the aorta (P < 0.05). Alizarin red and von Kossa staining reveal notable reductions in calcium salt deposits in VSMCs and aorta, respectively. Notably, the expression levels of BMP-2, PINP, OC, and BALP were substantially decreased in VSMCs (P < 0.05), underscoring ENPP1's role in impeding osteoblast-like transdifferentiation of VSMCs. Additionally, ENPP1 overexpression led to a significant increase in pyrophosphate (PPi) levels compared to control rats (P < 0.05). In conclusion, this study suggests that ENPP1 contributes to alleviating vascular calcification by elevating PPi levels and inhibiting the phenotypic transformation of VSMCs. These findings shed light on the potential therapeutic role of ENPP1 in mitigating vascular calcification-related complications.
Collapse
Affiliation(s)
- Xiujuan Wu
- Department of Nephrology, Shaoxing People’s Hospital, Shaoxing312000, Zhejiang Province, China
| | - Shuijuan Shen
- Department of Nephrology, Shaoxing People’s Hospital, Shaoxing312000, Zhejiang Province, China
| | - Jiaying Wu
- Department of Nephrology, Shaoxing People’s Hospital, Shaoxing312000, Zhejiang Province, China
| | - Shaorui Wu
- Department of Nephrology, Shaoxing People’s Hospital, Shaoxing312000, Zhejiang Province, China
| | - Shimi Wang
- Department of Nephrology, Shaoxing People’s Hospital, Shaoxing312000, Zhejiang Province, China
| | - Feng Di
- Department of Respiratory, Shaoxing People’s Hospital, Yuecheng District, No. 568 Zhongxing North Road, Shaoxing312000, Zhejiang Province, China
| |
Collapse
|
10
|
Yang S, Zeng Z, Yuan Q, Chen Q, Wang Z, Xie H, Liu J. Vascular calcification: from the perspective of crosstalk. MOLECULAR BIOMEDICINE 2023; 4:35. [PMID: 37851172 PMCID: PMC10584806 DOI: 10.1186/s43556-023-00146-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
Vascular calcification (VC) is highly correlated with cardiovascular disease morbidity and mortality, but anti-VC treatment remains an area to be tackled due to the ill-defined molecular mechanisms. Regardless of the type of VC, it does not depend on a single cell but involves multi-cells/organs to form a complex cellular communication network through the vascular microenvironment to participate in the occurrence and development of VC. Therefore, focusing only on the direct effect of pathological factors on vascular smooth muscle cells (VSMCs) tends to overlook the combined effect of other cells and VSMCs, including VSMCs-VSMCs, ECs-VMSCs, Macrophages-VSMCs, etc. Extracellular vesicles (EVs) are a collective term for tiny vesicles with a membrane structure that are actively secreted by cells, and almost all cells secrete EVs. EVs docked on the surface of receptor cells can directly mediate signal transduction or transfer their contents into the cell to elicit a functional response from the receptor cells. They have been proven to participate in the VC process and have also shown attractive therapeutic prospects. Based on the advantages of EVs and the ability to be detected in body fluids, they may become a novel therapeutic agent, drug delivery vehicle, diagnostic and prognostic biomarker, and potential therapeutic target in the future. This review focuses on the new insight into VC molecular mechanisms from the perspective of crosstalk, summarizes how multi-cells/organs interactions communicate via EVs to regulate VC and the emerging potential of EVs as therapeutic methods in VC. We also summarize preclinical experiments on crosstalk-based and the current state of clinical studies on VC-related measures.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaolin Zeng
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qing Yuan
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qian Chen
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hui Xie
- Department of Orthopaedics, Movement System Injury and Repair Research Centre, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
11
|
Lee S, Kalra K, Kashikar A, Redpath B, Bernheim A, Brewster L, Shaw L, Arya S. Evaluation of Lower Extremity Calcium Score as a Measure of Peripheral Arterial Disease Burden and Amputation Risk. Ann Vasc Surg 2023; 95:154-161. [PMID: 36889632 DOI: 10.1016/j.avsg.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/04/2023] [Accepted: 02/07/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND The ankle-brachial pressure index (ABI) and toe-brachial pressure index (TBI) are commonly used diagnostic tools for peripheral artery disease (PAD) that are unreliable in the presence of calcified vessels. In this study, we aimed to demonstrate the utility of the lower extremity calcium score (LECS) in addition to ABI and TBI in measuring disease burden and predicting the risk of amputation in patients with PAD. METHODS Patients who were evaluated in the vascular surgery clinic at Emory University for PAD and who underwent noncontrast computed tomography of the aorta and lower extremities were included in the study. Aortoiliac, femoral-popliteal, and tibial calcium scores were measured using the Agatston method. ABI and TBI that were obtained within 6 months of the computed tomography scan were noted and divided into categories of PAD severity. Associations between ABI, TBI, and LECS of each anatomic segment were evaluated. Univariate and multivariate ordinal regression analyses were performed to predict the outcome of amputation. Receiver operating characteristic analysis was performed to compare LECS with other variables in its ability to predict amputation. RESULTS Fifty patients included in the study cohort were divided into LECS quartiles, with 12-13 patients in each quartile. The highest quartile tended to be older (P = 0.016), had a higher percentage of diabetics (P = 0.034), and had a higher frequency of major amputations (P = 0.004) compared to the other quartiles. Patients in the highest quartile of tibial calcium score were more likely to have stage 3 chronic kidney disease (CKD) or greater (P = 0.011) and also had a higher frequency of amputation (P < 0.005) and mortality (P = 0.041). We found no significant association between each anatomic LECS and ABI/TBI categories. On univariate analysis, CKD (Odds Ratio [OR] 12.92 (95% CI 2.01 to 82.83), P = 0.007), diabetes mellitus (OR 5.47 (95% CI 1.27 to 23.64), P = 0.023), tibial calcium score (OR 6.62 (95% CI 1.79 to 24.54), P = 0.005), and total bilateral calcium score (OR 6.32 (95% CI 1.18 to 33.78), P = 0.031) were associated with increased risk of amputation. On multivariate stepwise ordinal regression, TBI and tibial calcium score were identified as important predictors of amputation, with hyperlipidemia and CKD increasing the overall prediction of the model. On Receiver operating characteristic analysis, the addition of the tibial calcium score (area under the curve 0.94, standard error 0.048) significantly improved the prediction of amputation compared to hyperlipidemia, CKD, and TBI alone (area under the curve 0.82, standard error 0.071, P = 0.022). CONCLUSIONS The addition of tibial calcium score to other known PAD risk factors may improve the prediction of amputation in patients with PAD.
Collapse
Affiliation(s)
- Sujin Lee
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Emory University School of Medicine, Atlanta, GA; Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Massachusetts General Hospital, Boston, MA; Surgical Service Line, Atlanta VA Medical Center, Decatur, GA; Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA.
| | - Kanika Kalra
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Emory University School of Medicine, Atlanta, GA; Surgical Service Line, Atlanta VA Medical Center, Decatur, GA; Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
| | - Aditi Kashikar
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Emory University School of Medicine, Atlanta, GA; Surgical Service Line, Atlanta VA Medical Center, Decatur, GA; Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
| | - Benjamin Redpath
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA; Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA; Palo Alto VA Medical Center, Palo Alto, CA; Surgical Services, Palo Alto VA Medical Center, Palo Alto, CA
| | - Adam Bernheim
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA; Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA; Palo Alto VA Medical Center, Palo Alto, CA; Surgical Services, Palo Alto VA Medical Center, Palo Alto, CA
| | - Luke Brewster
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Emory University School of Medicine, Atlanta, GA; Surgical Service Line, Atlanta VA Medical Center, Decatur, GA
| | - Leslee Shaw
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA
| | - Shipra Arya
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Palo Alto, CA; Palo Alto VA Medical Center, Palo Alto, CA; Surgical Services, Palo Alto VA Medical Center, Palo Alto, CA
| |
Collapse
|
12
|
Ibragimova AG, Stanishevskiy YM, Plakkhin AM, Zubko AV, Darvish NA, Koassary AK, Shindyapina AV. Comparative analysis of calcified soft tissues revealed shared deregulated pathways. Front Aging Neurosci 2023; 15:1131548. [PMID: 37441678 PMCID: PMC10335799 DOI: 10.3389/fnagi.2023.1131548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/18/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction Calcification of soft tissues is a common age-related pathology that primarily occurs within vascular tissue. The mechanisms underlying pathological calcification in humans and tissue specificity of the process is still poorly understood. Previous studies examined calcified tissues on one to one basis, thus preventing comparison of deregulated pathways across tissues. Purpose This study aimed to establish common and tissue-specific changes associated with calcification in aorta, artery tibial, coronary artery and pituitary gland in subjects from the Genotype-Tissue Expression (GTEx) dataset using its RNA sequencing and histological data. Methods We used publicly available data from the GTEx database https://gtexportal.org/home/aboutGTEx. All GTEx tissue samples were derived by the GTEx consorcium from deceased donors, with age from 20 to 79, both men and women. GTEx study authorization was obtained via next-of-kin consent for the collection and banking of de-identified tissue samples for scientific research. Hematoxylin and eosin (H&E) staining of arteries were manually graded based on the presence of calcification on a scale from zero to four, where zero designates absence of calcification and four designates severe calcification. Samples with fat contamination and mislabeled tissues were excluded, which left 430 aorta, 595 artery tibial, 124 coronary artery, and 283 pituitary samples for downstream gene expression analysis. Transcript levels of protein-coding genes were associated with calcification grade using sex, age bracket and cause of death as covariates, and tested for pathway enrichment using gene set enrichment analysis. Results We identified calcification deposits in 28 (6.5%) aortas, 121 (20%), artery tibials, 54 (43%), coronary arteries, and 24 (8%) pituitary glands of GTEx subjects. We observed an age-dependent increase in incidence of calcification in all vascular tissues, but not in pituitary. Subjects with calcification in the artery tibial were significantly more likely to have calcification in the coronary artery (OR = 2.56, p = 6.3e-07). Markers of calcification previously established in preclinical and in vitro studies, e.g., BMP2 and RUNX2, were deregulated in the calcified tibial and coronary arteries, confirming the relevance of these genes to human pathology. Differentially expressed genes associated with calcification poorly overlapped across tissues suggesting tissue-specific nuances in mechanisms of calcification. Nevertheless, calcified arteries unanimously down-regulated pathways of intracellular transport and up-regulated inflammatory pathways suggesting these as universal targets for pathological calcification. In particular, PD-1 and PD-L1 genes were up-regulated in calcified tissues but not in the blood of the same subjects, suggesting that localized inflammation contributes to pathological calcification. Conclusion Pathological calcification is a prevalent disease of aging that shares little changes in expression in individual genes across tissues. However, our analysis suggests that it potentially can be targeted by alleviating local inflammation of soft tissues.
Collapse
Affiliation(s)
| | | | | | | | - Nidal Akhmedovich Darvish
- Bakoulev National Medical Research Center for Cardiovascular Surgery, Russian Federation, Moscow, Russia
| | - Anton Karenovich Koassary
- Bakoulev National Medical Research Center for Cardiovascular Surgery, Russian Federation, Moscow, Russia
| | - Anastasia V. Shindyapina
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Retro Biosciences Inc., Redwood City, CA, United States
| |
Collapse
|
13
|
Dai Z, Zhang X. Pathophysiology and Clinical Impacts of Chronic Kidney Disease on Coronary Artery Calcification. J Cardiovasc Dev Dis 2023; 10:jcdd10050207. [PMID: 37233174 DOI: 10.3390/jcdd10050207] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
The global prevalence of chronic kidney disease (CKD) has increased in recent years. Adverse cardiovascular events have become the main cause of life-threatening events in patients with CKD, and vascular calcification is a risk factor for cardiovascular disease. Vascular calcification, especially coronary artery calcification, is more prevalent, severe, rapidly progressive, and harmful in patients with CKD. Some features and risk factors are unique to vascular calcification in patients with CKD; the formation of vascular calcification is not only influenced by the phenotypic transformation of vascular smooth muscle cells, but also by electrolyte and endocrine dysfunction, uremic toxin accumulation, and other novel factors. The study on the mechanism of vascular calcification in patients with renal insufficiency can provide a basis and new target for the prevention and treatment of this disease. This review aims to illustrate the impact of CKD on vascular calcification and to discuss the recent research data on the pathogenesis and factors involved in vascular calcification, mainly focusing on coronary artery calcification, in patients with CKD.
Collapse
Affiliation(s)
- Zhuoming Dai
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xiangyu Zhang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
14
|
Huang A, Li L, Liu X, Lian Q, Guo G, Xu T, Lu X, Ma L, Ma H, Yu Y, Yao L. Hedgehog signaling is a potential therapeutic target for vascular calcification. Gene 2023; 872:147457. [PMID: 37141952 DOI: 10.1016/j.gene.2023.147457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/07/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) suffered from vascular calcification (VC), one major contributor for their increased mortality rate. Hedgehog (Hh) signaling plays a crucial role in physiological bone mineralization and is associated with several cardiovascular diseases. However, the molecular changes underlying VC is ill defined and it remains unclear whether Hh signaling intervention affects VC. METHODS We constructed human primary vascular smooth muscle cell (VSMC) calcification model and performed RNA sequencing. Alizarin red staining and calcium content assay were conducted to identify the occurrence of VC. Three different R packages were applied to determine differentially expressed genes (DEGs). Enrichment analysis and protein-protein interaction (PPI) network analysis were carried out to explore the biological roles of DEGs. qRT-PCR assay was then applied to validate the expression of key genes. By using Connectivity Map (CMAP) analysis, several small molecular drugs targeting these key genes were obtained, including SAG (Hedgehog signaling activator) and cyclopamine (CPN) (Hedgehog signaling inhibitor), which were subsequently used to treat VSMC. RESULTS Obvious Alizarin red staining and increased calcium content identified the occurrence of VC. By integrating results from three R packages, we totally obtained 166 DEGs (86 up-regulated and 80 down-regulated), which were significantly enriched in ossification, osteoblast differentiation, and Hh signaling. PPI network analysis identified 10 key genes and CMAP analysis predicted several small molecular drugs targeting these key genes including chlorphenamine, isoeugenol, CPN and phenazopyridine. Notably, our in vitro experiment showed that SAG markedly alleviated VSMC calcification, whereas CPN significantly exacerbated VC. CONCLUSIONS Our research provided deeper insight to the pathogenesis of VC and indicated that targeting Hh signaling pathway may represent a potential and effective therapy for VC.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Lu Li
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Xiaoxu Liu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Qiuting Lian
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Guangying Guo
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Ling Ma
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Haiying Ma
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Yanqiu Yu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China; Shenyang Engineering Technology R&D Center of Cell Therapy Co. LTD., Shenyang 110169, China.
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
15
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
16
|
GDF11 Is a Novel Protective Factor Against Vascular Calcification. J Cardiovasc Pharmacol 2022; 80:852-860. [PMID: 36027600 DOI: 10.1097/fjc.0000000000001357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/31/2022] [Indexed: 12/13/2022]
Abstract
ABSTRACT Vascular calcification (VC) occurs via an active cell-mediated process, which involves osteogenic differentiation, apoptosis, and phenotypic transformation of vascular smooth muscle cells (VSMCs). As a member of the transforming growth factor-β family, growth differentiation factor 11 (GDF11) can inhibit apoptosis and osteogenic differentiation and maintain the stability of atherosclerotic plaques. In this study, coronary artery calcium score (CACS) of participants with GDF11 measurements was measured using computed tomography angiography and was scored according to the Agatston score. β-glycerophosphate (10 mM), dexamethasone (100 nM), and l -ascorbic acid (50 µg/mL) [osteogenic medium (OM)] were used to induce calcification of human aortic smooth muscle cells. We found that CACS was negatively correlated with serum GDF11 levels in patients and GDF11 was a strong predictor of elevated CACS (OR = 0.967, 95% CI: 0.945-0.991; P = 0.006), followed by age (OR = 1.151, 95% CI: 1.029-1.286; P = 0.014), triglycerides (OR = 4.743, 95% CI: 1.170-19.236; P = 0.029), C-reactive protein (OR = 1.230, 95% CI: 1.010-1.498; P = 0.04), and hypertension (OR = 7.264, 95% CI: 1.099-48.002; P = 0.04). Furthermore, exogenous GDF11 inhibited OM-induced calcification by inhibiting osteogenic differentiation, the phenotypic transformation and apoptosis of human aortic smooth muscle cells. Our study demonstrates that GDF11 plays a crucial role in reducing vascular calcification and serves as a potential intervention target to vascular calcification.
Collapse
|
17
|
Luo F, Guo W, Liu W. Exosomes derived from bone marrow mesenchymal stem cells inhibit human aortic vascular smooth muscle cells calcification via the miR-15a/15b/16/NFATc3/OCN axis. Biochem Biophys Res Commun 2022; 635:65-76. [DOI: 10.1016/j.bbrc.2022.09.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 12/15/2022]
|
18
|
Nazifova-Tasinova NF, Atanasov AA, Pasheva MG, Yotov YT, Gerova DI, Vankova DG, Todorova MN, Ivanova DG, Kiselova-Kaneva YD, Galunska BT. Circulating uncarboxylated matrix Gla protein in patients with atrial fibrillation or heart failure with preserved ejection fraction. Arch Physiol Biochem 2022; 128:1619-1629. [PMID: 32620059 DOI: 10.1080/13813455.2020.1786130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 10/23/2022]
Abstract
CONTEXT Circulating uncarboxylated matrix Gla protein (ucMGP) is possibly related to coronary arterial calcification (CAC) in cardiovascular disease (CVD) patients. OBJECTIVE We aimed to evaluate the relationships between circulating ucMGP, CVD pathology and CAC and its interplay with CVD risk factors. MATERIALS AND METHODS ucMGP was measured in 99 CVD-patients. CAC score was determined by multislice computed tomography. Circulating ucMGP, uncarboxylated (ucOC) and carboxylated osteocalcin (cOC) were assayed by ELISA kits. Vitamin-K status was evaluated by ucOC/cOC ratio. RESULTS A tendency for decreased ucMGP was observed for CAC ≥ 100 AU vs. CAC = 1-99 AU after exclusion of the patients on vitamin K-antagonist anticoagulants. Significant inverse correlations between ucMGP and vitamin-K status were indicated for the entire cohort and according to CAC score. Significant associations were found between ucMGP and risk factors for CVD. CONCLUSION Circulating ucMGP may reflect certain stages of CVD and CAC. Future studies are needed to clarify its role as potential biomarker.
Collapse
Affiliation(s)
| | | | - Milena Gincheva Pasheva
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical university of Varna, Varna, Bulgaria
| | - Yoto Trifonov Yotov
- First Department of Internal Diseases, Medical university of Varna, Varna, Bulgaria
| | - Daniela Ivanova Gerova
- Department of General Medicine and Clinical Laboratory, Medical university of Varna, Varna, Bulgaria
| | - Deyana Georgieva Vankova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical university of Varna, Varna, Bulgaria
| | - Miglena Nikolaeva Todorova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical university of Varna, Varna, Bulgaria
| | - Diana Georgieva Ivanova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical university of Varna, Varna, Bulgaria
| | | | - Bistra Tzaneva Galunska
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Medical university of Varna, Varna, Bulgaria
| |
Collapse
|
19
|
Liu K, Yu Y, Yuan Y, Xu X, Lei W, Niu R, Shen M, Zhou L, Peng R, Wang Q, Yang H, Guo H, Ge Y, Liu G, He M, Wu T, Zhang X. Elevated Levels of Serum Alkaline Phosphatase are Associated with Increased Risk of Cardiovascular Disease: A Prospective Cohort Study. J Atheroscler Thromb 2022:63646. [PMID: 36261365 DOI: 10.5551/jat.63646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023] Open
Abstract
AIM We aimed to investigate the associations of serum alkaline phosphatase (ALP) levels with incident cardiovascular disease (CVD), coronary heart disease (CHD), and stroke, as well as their subtypes, among men and women in a prospective cohort study. METHODS A total of 11,408 men and 14,981 women were included to evaluate the associations between ALP levels and incident CVD. Participants were divided into four groups according to the quartiles of serum ALP levels in men and women separately. Cox proportional hazard models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS During an average follow-up of 7.3 years, 7,015 incident CVDs (5,561 CHDs and 1,454 strokes) were documented. After adjustments for age, body mass index, smoking status, drinking status, diabetes, hyperlipidemia, hypertension, physical activity, aspirin usage, anticoagulants usage, menopausal status (women only), family history of CVD, estimated glomerular filtration rate, white blood cell counts, and admission batch and comparing the lowest quartile of ALP, the adjusted HRs (95% CIs) of participants in the highest quartile were 1.22 (1.11-1.34) for CVD, 1.14 (1.02-1.28) for CHD, 1.43 (1.18-1.73) for stroke, 1.31 (1.09-1.57) for acute coronary syndrome (ACS), 1.37 (1.11-1.70) for ischemic stroke, and 1.75 (1.10-2.79) for hemorrhagic stroke in men and 1.12 (1.01-1.23) for CVD, 1.10 (0.99-1.23) for CHD, 1.18 (0.92-1.51) for stroke, 1.23 (1.03-1.47) for ACS, 1.10 (0.83-1.45) for ischemic stroke, and 1.54 (0.90-2.65) for hemorrhagic stroke in women. The ALP-CVD associations remained significant even within the normal ranges of ALP levels (40-150 U/L). Moreover, linear dose-response relationships were found between ALP levels and incident CVD. CONCLUSIONS Higher ALP levels, even within the normal range, were significantly associated with increased risks of CVD, in a dose-dependent manner. These findings suggested that regular monitoring of ALP levels may help in improving the early identification of the population at higher CVD risk.
Collapse
Affiliation(s)
- Kang Liu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
- School of Public Health, Guangzhou Medical University
| | - Yanqiu Yu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Yu Yuan
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Xuedan Xu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Wenhui Lei
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Rundong Niu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Miaoyan Shen
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Lue Zhou
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Rong Peng
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Qiuhong Wang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Handong Yang
- Department of Cardiovascular Diseases, Sinopharm Dongfeng General Hospital, Hubei University of Medicine
| | - Huan Guo
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Yang Ge
- School of Public Health, Shanghai Jiaotong University School of Medicine
| | - Gang Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology
| | - Meian He
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Tangchun Wu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College
| |
Collapse
|
20
|
Biruete A, Metzger CE, Chen NX, Swallow EA, Vrabec C, Clinkenbeard EL, Stacy AJ, Srinivasan S, O'Neill K, Avin KG, Allen MR, Moe SM. Effects of ferric citrate and intravenous iron sucrose on markers of mineral, bone, and iron homeostasis in a rat model of CKD-MBD. Nephrol Dial Transplant 2022; 37:1857-1867. [PMID: 35482713 PMCID: PMC9494145 DOI: 10.1093/ndt/gfac162] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Anemia and chronic kidney disease-mineral and bone disorder (CKD-MBD) are common and begin early in CKD. Limited studies have concurrently compared the effects of ferric citrate (FC) versus intravenous (IV) iron on CKD-MBD and iron homeostasis in moderate CKD. METHODS We tested the effects of 10 weeks of 2% FC versus IV iron sucrose in rats with moderate CKD (Cy/+ male rat) and untreated normal (NL) littermates. Outcomes included a comprehensive assessment of CKD-MBD, iron homeostasis and oxidative stress. RESULTS CKD rats had azotemia, elevated phosphorus, parathyroid hormone and fibroblast growth factor-23 (FGF23). Compared with untreated CKD rats, treatment with FC led to lower plasma phosphorus, intact FGF23 and a trend (P = 0.07) toward lower C-terminal FGF23. FC and IV iron equally reduced aorta and heart calcifications to levels similar to NL animals. Compared with NL animals, CKD animals had higher bone turnover, lower trabecular volume and no difference in mineralization; these were unaffected by either iron treatment. Rats treated with IV iron had cortical and bone mechanical properties similar to NL animals. FC increased the transferrin saturation rate compared with untreated CKD and NL rats. Neither iron treatment increased oxidative stress above that of untreated CKD. CONCLUSIONS Oral FC improved phosphorus homeostasis, some iron-related parameters and the production and cleavage of FGF23. The intermittent effect of low-dose IV iron sucrose on cardiovascular calcification and bone should be further explored in moderate-advanced CKD.
Collapse
Affiliation(s)
- Annabel Biruete
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Nutrition and Dietetics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Corinne E Metzger
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Neal X Chen
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth A Swallow
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Curtis Vrabec
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- College of Osteopathic Medicine, Marian University, Indianapolis, IN, USA
| | - Erica L Clinkenbeard
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander J Stacy
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shruthi Srinivasan
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kalisha O'Neill
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith G Avin
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Physical Therapy, Indiana University School of Health and Human Sciences, Indiana University, Indianapolis, IN, USA
| | - Matthew R Allen
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Affairs Medical Center, Indianapolis, IN, IN, USA
| | - Sharon M Moe
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Affairs Medical Center, Indianapolis, IN, IN, USA
| |
Collapse
|
21
|
Villa-Bellosta R. Role of the extracellular ATP/pyrophosphate metabolism cycle in vascular calcification. Purinergic Signal 2022:10.1007/s11302-022-09867-1. [PMID: 35511317 DOI: 10.1007/s11302-022-09867-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/19/2022] [Indexed: 10/18/2022] Open
Abstract
Conventionally, ATP is considered to be the principal energy source in cells. However, over the last few years, a novel role for ATP as a potent extracellular signaling molecule and the principal source of extracellular pyrophosphate, the main endogenous inhibitor of vascular calcification, has emerged. A large body of evidence suggests that two principal mechanisms are involved in the initiation and progression of ectopic calcification: high phosphate concentration and pyrophosphate deficiency. Pathologic calcification of cardiovascular structures, or vascular calcification, is a feature of several genetic diseases and a common complication of chronic kidney disease, diabetes, and aging. Previous studies have shown that the loss of function of several enzymes and transporters involved in extracellular ATP/pyrophosphate metabolism is associated with vascular calcification. Therefore, pyrophosphate homeostasis should be further studied to facilitate the design of novel therapeutic approaches for ectopic calcification of cardiovascular structures, including strategies to increase pyrophosphate concentrations by targeting the ATP/pyrophosphate metabolism cycle.
Collapse
Affiliation(s)
- Ricardo Villa-Bellosta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av Barcelona, Campus Vida, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain. .,Department of Biochemistry and Molecular Biology, Universidade de Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain.
| |
Collapse
|
22
|
Stremke ER, Trevino L, Doshi S, Moorthi RN, Hill Gallant KM, Moe SM. Postdialysis serum phosphate equilibrium in hemodialysis patients on a controlled diet and no binders. Hemodial Int 2022; 26:255-263. [PMID: 34964250 PMCID: PMC9096807 DOI: 10.1111/hdi.12999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Studies evaluating the change in serum phosphate post hemodialysis (HD) demonstrate an initial decline during dialysis but a rebound post dialysis. However, previous studies were done on usual diet and phosphate binders, with limited number of blood draws, confounding conclusions. We determined serum phosphate reduction, rebound, and equilibrium over 48 h in HD patients consuming a controlled, low phosphorus diet without binders. METHODS Serum phosphate (mg/dL) was analyzed before and after a HD treatment and frequently during the ensuing 48 h intradialytic period in the clinical research unit. Thirteen subjects were enrolled and had been off phosphate binders for 10 days and consumed a standardized low phosphate (900 mg/day) diet for 3 weeks prior to the assessments. Linear regression was used to determine relationships between the pre-HD serum phosphate, decline post-HD (post-HD drop); and a 48 h area under curve (AUC) using the trapezoidal method as a measure of overall phosphate levels from the end of dialysis to 48 h post dialysis. Repeated Measures ANOVA with Dunnett's posthoc test was used to determine rebound. RESULTS Five of 13 subjects returned to >90% of their pre-HD serum phosphate within the first 24 h post-HD, and serum phosphate was 94 ± 0.11% (range 63%-113%) by 48 h after the completion of HD. The 48 h AUC of serum phosphate during the interdialytic period was correlated with both pre dialysis phosphorus (r = 0.85; p = 0.0002) and the pre-post drop in serum phosphate during dialysis (r = 0.69; p = 0.0085). In contrast, the net ultrafiltration was not related to the 48 h AUC of serum phosphorus (r = 0.20; p = 0.51). CONCLUSIONS In hemodialysis patients on standard low phosphorus diet and no phosphate binders, the interdialytic serum phosphorus level, assessed as AUC, is determined by the pre dialysis phosphorus and net-change in serum phosphorus during the dialysis treatment, but not the ultrafiltration volume [Correction added on 25 January, after first online publication: In the last sentence of the Abstract, the word "potassium" has been replaced with "phosphorus" to improve accuracy.].
Collapse
Affiliation(s)
- Elizabeth R Stremke
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
- Indiana Clinical and Translational Science Institute, Indiana University, Indianapolis, Indiana, USA
| | - Laurie Trevino
- Indiana Clinical and Translational Science Institute, Indiana University, Indianapolis, Indiana, USA
| | - Simit Doshi
- Department of Medicine/Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ranjani N Moorthi
- Department of Medicine/Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kathleen M Hill Gallant
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, Minnesota, USA
| | - Sharon M Moe
- Indiana Clinical and Translational Science Institute, Indiana University, Indianapolis, Indiana, USA
- Department of Medicine/Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
23
|
Li T, Yu H, Zhang D, Feng T, Miao M, Li J, Liu X. Matrix Vesicles as a Therapeutic Target for Vascular Calcification. Front Cell Dev Biol 2022; 10:825622. [PMID: 35127686 PMCID: PMC8814528 DOI: 10.3389/fcell.2022.825622] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/04/2022] [Indexed: 01/01/2023] Open
Abstract
Vascular calcification (VC) is linked to an increased risk of heart disease, stroke, and atherosclerotic plaque rupture. It is a cell-active process regulated by vascular cells rather than pure passive calcium (Ca) deposition. In recent years, extracellular vesicles (EVs) have attracted extensive attention because of their essential role in the process of VC. Matrix vesicles (MVs), one type of EVs, are especially critical in extracellular matrix mineralization and the early stages of the development of VC. Vascular smooth muscle cells (VSMCs) have the potential to undergo phenotypic transformation and to serve as a nucleation site for hydroxyapatite crystals upon extracellular stimulation. However, it is not clear what underlying mechanism that MVs drive the VSMCs phenotype switching and to result in calcification. This article aims to review the detailed role of MVs in the progression of VC and compare the difference with other major drivers of calcification, including aging, uremia, mechanical stress, oxidative stress, and inflammation. We will also bring attention to the novel findings in the isolation and characterization of MVs, and the therapeutic application of MVs in VC.
Collapse
Affiliation(s)
- Tiantian Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hongchi Yu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Demao Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Tang Feng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Michael Miao
- Division of Oral & Craniofacial Health Sciences, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, United States
| | - Jianwei Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jianwei Li, ; Xiaoheng Liu,
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Jianwei Li, ; Xiaoheng Liu,
| |
Collapse
|
24
|
Ramos AP, Sebinelli HG, Ciancaglini P, Rosato N, Mebarek S, Buchet R, Millán JL, Bottini M. The functional role of soluble proteins acquired by extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e34. [PMID: 38938684 PMCID: PMC11080634 DOI: 10.1002/jex2.34] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed nanosized particles released by all cell types during physiological as well as pathophysiological processes to carry out diverse biological functions, including acting as sources of cellular dumping, signalosomes and mineralisation nanoreactors. The ability of EVs to perform specific biological functions is due to their biochemical machinery. Among the components of the EVs' biochemical machinery, surface proteins are of critical functional significance as they mediate the interactions of EVs with components of the extracellular milieu, the extracellular matrix and neighbouring cells. Surface proteins are thought to be native, that is, pre-assembled on the EVs' surface by the parent cells before the vesicles are released. However, numerous pieces of evidence have suggested that soluble proteins are acquired by the EVs' surface from the extracellular milieu and further modulate the biological functions of EVs during innate and adaptive immune responses, autoimmune disorders, complement activation, coagulation, viral infection and biomineralisation. Herein, we will describe the methods currently used to identify the EVs' surface proteins and discuss recent knowledge on the functional relevance of the soluble proteins acquired by EVs.
Collapse
Affiliation(s)
- Ana Paula Ramos
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Heitor Gobbi Sebinelli
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Pietro Ciancaglini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Nicola Rosato
- Dipartimento di Medicina SperimentaleUniversita’ di Roma “Tor Vergata”RomeItaly
| | - Saida Mebarek
- ICBMS UMR CNRS 5246UFR BiosciencesUniversité Lyon 1Villeurbanne CedexFrance
| | - Rene Buchet
- ICBMS UMR CNRS 5246UFR BiosciencesUniversité Lyon 1Villeurbanne CedexFrance
| | | | - Massimo Bottini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
- Sanford Burnham PrebysLa JollaCaliforniaUSA
| |
Collapse
|
25
|
Sivakumar B, Kurian GA. Mitochondria and traffic-related air pollution linked coronary artery calcification: exploring the missing link. REVIEWS ON ENVIRONMENTAL HEALTH 2021; 36:545-563. [PMID: 34821115 DOI: 10.1515/reveh-2020-0127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/04/2021] [Indexed: 06/13/2023]
Abstract
The continuing increase in the exposure to Traffic-related air pollution (TRAP) in the general population is predicted to result in a higher incidence of non-communicable diseases like cardiovascular disease. The chronic exposure of air particulate matter from TRAP upon the vascular system leads to the enhancement of deposition of calcium in the vasculature leading to coronary artery calcification (CAC), triggered by inflammatory reactions and endothelial dysfunction. This calcification forms within the intimal and medial layers of vasculature and the underlying mechanism that connects the trigger from TRAP is not well explored. Several local and systemic factors participate in this active process including inflammatory response, hyperlipidemia, presence of self-programmed death bodies and high calcium-phosphate concentrations. These factors along with the loss of molecules that inhibit calcification and circulating nucleation complexes influence the development of calcification in the vasculature. The loss of defense to prevent osteogenic transition linked to micro organelle dysfunction that includes deteriorated mitochondria, elevated mitochondrial oxidative stress, and defective mitophagy. In this review, we examine the contributory role of mitochondria involved in the mechanism of TRAP linked CAC development. Further we examine whether TRAP is an inducer or trigger for the enhanced progression of CAC.
Collapse
Affiliation(s)
- Bhavana Sivakumar
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Gino A Kurian
- Vascular Biology Lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| |
Collapse
|
26
|
Regulation of MDM2 E3 ligase-dependent vascular calcification by MSX1/2. Exp Mol Med 2021; 53:1781-1791. [PMID: 34845330 PMCID: PMC8639964 DOI: 10.1038/s12276-021-00708-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/24/2021] [Accepted: 10/06/2021] [Indexed: 11/27/2022] Open
Abstract
Vascular calcification increases morbidity and mortality in patients with cardiovascular and renal diseases. Previously, we reported that histone deacetylase 1 prevents vascular calcification, whereas its E3 ligase, mouse double minute 2 homolog (MDM2), induces vascular calcification. In the present study, we identified the upstream regulator of MDM2. By utilizing cellular models and transgenic mice, we confirmed that E3 ligase activity is required for vascular calcification. By promoter analysis, we found that both msh homeobox 1 (Msx1) and msh homeobox 2 (Msx2) bound to the MDM2 promoter region, which resulted in transcriptional activation of MDM2. The expression levels of both Msx1 and Msx2 were increased in mouse models of vascular calcification and in calcified human coronary arteries. Msx1 and Msx2 potentiated vascular calcification in cellular and mouse models in an MDM2-dependent manner. Our results establish a novel role for MSX1/MSX2 in the transcriptional activation of MDM2 and the resultant increase in MDM2 E3 ligase activity during vascular calcification. The identification of a signaling pathway involved in triggering vascular calcification, the deposition of calcium phosphate crystals in blood vessels, could inform new therapeutic interventions for related cardiovascular complications. Vascular calcification causes significant complications in patients with metabolic syndrome, renal failure, or cardiovascular disease. In their previous work, Hyun Kook and Duk-Hwa Kwon at Chonnam National University Medical School, Jeollanamdo, Republic of Korea, and coworkers demonstrated that the E3 ligase activity of a protein called MDM2 induces calcification. Now, following further mouse trials, the team have identified an upstream signaling pathway involving several development proteins such as MSX1 and MSX2 which activate MDM2. The activation of this signaling axis leads to the degradation of a key protein that would otherwise prevent calcification. The results may provide a platform for novel therapies targeting the condition.
Collapse
|
27
|
Li W, Feng W, Su X, Luo D, Li Z, Zhou Y, Zhu Y, Zhang M, Chen J, Liu B, Huang H. SIRT6 protects vascular smooth muscle cell from osteogenic transdifferentiation via Runx2 in chronic kidney disease. J Clin Invest 2021; 132:150051. [PMID: 34793336 PMCID: PMC8718147 DOI: 10.1172/jci150051] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/12/2021] [Indexed: 12/03/2022] Open
Abstract
Vascular calcification (VC) is regarded as an important pathological change lacking effective treatment and associated with high mortality. Sirtuin 6 (SIRT6) is a member of the Sirtuin family, a class III histone deacetylase and a key epigenetic regulator. SIRT6 has a protective role in patients with chronic kidney disease (CKD). However, the exact role and molecular mechanism of SIRT6 in VC in patients with CKD remain unclear. Here, we demonstrated that SIRT6 was markedly downregulated in peripheral blood mononuclear cells (PBMCs) and in the radial artery tissue of patients with CKD with VC. SIRT6-transgenic (SIRT6-Tg) mice showed alleviated VC, while vascular smooth muscle cell–specific (VSMC-specific) SIRT6 knocked-down mice showed severe VC in CKD. SIRT6 suppressed the osteogenic transdifferentiation of VSMCs via regulation of runt-related transcription factor 2 (Runx2). Coimmunoprecipitation (co-IP) and immunoprecipitation (IP) assays confirmed that SIRT6 bound to Runx2. Moreover, Runx2 was deacetylated by SIRT6 and further promoted nuclear export via exportin 1 (XPO1), which in turn caused degradation of Runx2 through the ubiquitin-proteasome system. These results demonstrated that SIRT6 prevented VC by suppressing the osteogenic transdifferentiation of VSMCs, and as such targeting SIRT6 may be an appealing therapeutic target for VC in CKD.
Collapse
Affiliation(s)
- Wenxin Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weijing Feng
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Shenzhen, China
| | - Xiaoyan Su
- Department of Nephropathy, Tungwah Hospital of Sun Yat-Sen University, Dongguan, China
| | - Dongling Luo
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhibing Li
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongqiao Zhou
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongjun Zhu
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Mengbi Zhang
- Department of Nephropathy, Tungwah Hospital of Sun Yat-Sen University, Dongguan, China
| | - Jie Chen
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Baohua Liu
- Shenzhen University Health Science Center, ShenZhen University, Shenzhen, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
28
|
Erez D, Fanadka F, Benchetrit S, Cohen-Hagai K. The Combined Prognostic Significance of Alkaline Phosphatase and Intracranial Arterial Calcifications in Hemodialysis Patients. Am J Nephrol 2021; 52:763-770. [PMID: 34569494 DOI: 10.1159/000518399] [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: 05/01/2021] [Accepted: 07/06/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The prevalence of intracranial arterial calcification (ICAC) in maintenance hemodialysis (MHD) patients is about 90%, and its severity is correlated with age, hemodialysis vintage, and mineral bone disease. Elevated concentrations of calcium and phosphorus are not sufficient for medial calcification because of inhibition by pyrophosphate. Alkaline phosphatase (ALP) promotes calcification by hydrolyzing extracellular pyrophosphate. Epigenetic mechanisms involving ALP inhibition by apabetalone were investigated as a potential target for preventing vascular calcifications (VCs). This study assessed the combined impact of VCs and elevated serum ALP on mortality among chronic HD patients. METHODS VCs represented by ICAC were measured simultaneously with mineral bone disease parameters including serum ALP of MHD patients who underwent noncontrast brain computed tomography from 2015 to 2018 in our institution. RESULTS This retrospective study included 150 MHD patients (mean age 71.3 ± 12.1 years, 60.1% male). Of the total cohort, 12 (7.8%) had no brain calcifications and 69 (45.1%) had multiple intracranial calcifications. Considering the patients with normal ALP and no calcification as the reference group yielded adjusted odds ratios for all-cause mortality of 4.6 (95% CI: 1.7-12.7) among patients with brain calcifications and normal ALP (p = 0.003) and odds ratios for all-cause mortality of 6.1 (95% CI: 2.1-17.7) among patients with brain calcifications and elevated ALP (p= 0.001). CONCLUSION We found an independent association between ICAC and the risk of death among MHD patients. The combined effect of ICAC and elevated ALP was associated with a higher odds ratio for all-cause mortality in MHD patients and may contribute to the risk stratification of these patients.
Collapse
Affiliation(s)
- Daniel Erez
- Department of Internal Medicine D, Meir Medical Center, Kfar Saba, Israel
| | - Feda Fanadka
- Department of Radiology, Meir Medical Center, Kfar Saba, Israel
| | - Sydney Benchetrit
- Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Keren Cohen-Hagai
- Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
29
|
Campbell VK, Gately RP, Krishnasamy R, Burg D, Robertson GR, Gray NA. Midkine and chronic kidney disease-associated multisystem organ dysfunctions. Nephrol Dial Transplant 2021; 36:1577-1584. [PMID: 32542315 DOI: 10.1093/ndt/gfaa084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 02/18/2020] [Indexed: 12/18/2022] Open
Abstract
Chronic kidney disease (CKD) is a progressive multisystem condition with yet undefined mechanistic drivers and multiple implicated soluble factors. If identified, these factors could be targeted for therapeutic intervention for a disease that currently lacks specific treatment. There is increasing preclinical evidence that the heparin/endothelial glycocalyx-binding molecule midkine (MK) has a pathological role in multiple CKD-related, organ-specific disease processes, including CKD progression, hypertension, vascular and cardiac disease, bone disease and CKD-related cancers. Concurrent with this are studies documenting increases in circulating and urine MK proportional to glomerular filtration rate (GFR) loss in CKD patients and evidence that administering soluble MK reverses the protective effects of MK deficiency in experimental kidney disease. This review summarizes the growing body of evidence supporting MK's potential role in driving CKD-related multisystem disease, including MK's relationship with the endothelial glycocalyx, the deranged MK levels and glycocalyx profile in CKD patients and a proposed model of MK organ interplay in CKD disease processes and highlights the importance of ongoing research into MK's potential as a therapeutic target.
Collapse
Affiliation(s)
- Victoria K Campbell
- Renal Unit, Sunshine Coast University Hospital, Birtinya, Queensland, Australia.,University of Queensland, St Lucia, Queensland, Australia.,Intensive Care Unit, Sunshine Coast University Hospital, Birtinya, Queensland, Australia
| | - Ryan P Gately
- Renal Unit, Sunshine Coast University Hospital, Birtinya, Queensland, Australia
| | - Rathika Krishnasamy
- Renal Unit, Sunshine Coast University Hospital, Birtinya, Queensland, Australia.,University of Queensland, St Lucia, Queensland, Australia
| | | | | | - Nicholas A Gray
- Renal Unit, Sunshine Coast University Hospital, Birtinya, Queensland, Australia.,Sunshine Coast Health Institute, Birtinya, Queensland, Australia.,University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| |
Collapse
|
30
|
Liu Q, Yu L, Yin X, Ye J, Li S. Correlation Between Soluble Klotho and Vascular Calcification in Chronic Kidney Disease: A Meta-Analysis and Systematic Review. Front Physiol 2021; 12:711904. [PMID: 34483963 PMCID: PMC8414804 DOI: 10.3389/fphys.2021.711904] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/14/2021] [Indexed: 11/29/2022] Open
Abstract
Background: The correlation between soluble Klotho (sKlotho) level and vascular calcification (VC) in patients with chronic kidney disease (CKD) remains controversial. Using meta-analysis, we aimed to address this controversy and assess the feasibility of applying sKlotho as a biomarker for VC. Methods: Medical electronic databases were thoroughly searched for eligible publications on the association between sKlotho level and VC in CKD patients. Effectors, including correlation coefficients (r), odds ratios (ORs), hazard ratio (HR) or β-values, and 95% confidence intervals (CIs) were extracted and combined according to study design or effector calculation method. Pooled effectors were generated using both random-effects models and fixed-effects models according to I 2-value. Origin of heterogeneity was explored by sensitivity analysis and subgroup analysis. Results: Ten studies with 1,204 participants from a total of 1,199 publications were eligible and included in this meta-analysis. The combined correlation coefficient (r) was [-0.33 (-0.62, -0.04)] with significant heterogeneity (I 2 = 89%, p < 0.001) based on Spearman correlation analysis, and this significant association was also demonstrated in subgroups. There was no evidence of publication bias. The combined OR was [3.27 (1.70, 6.30)] with no evidence of heterogeneity (I 2 = 0%, p = 0.48) when sKlotho was treated as a categorical variable or [1.05 (1.01, 1.09)] with moderate heterogeneity (I 2 = 63%, p = 0.10) when sKlotho was treated as a continuous variable based on multivariate logistic regression. No significant association was observed and the pooled OR was [0.29 (0.01, 11.15)] with high heterogeneity (I 2 = 96%, p < 0.001) according to multivariate linear regression analysis. There was an inverse association between sKlotho and parathyroid hormone levels. The combined coefficient (r) was [-0.20 (-0.40, -0.01)] with significant heterogeneity (I 2 = 86%, p < 0.001), and without obvious publication bias. No significant association was found between sKlotho and calcium or phosphate levels. Conclusion: There exists a significant association between decreased sKlotho level and increased risk of VC in CKD patients. This raises the possibility of applying sKlotho as a biomarker for VC in CKD populations. Large, prospective, well-designed studies or interventional clinical trials are required to validate our findings.
Collapse
Affiliation(s)
- QiFeng Liu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - LiXia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - XiaoYa Yin
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - JianMing Ye
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - ShaSha Li
- Clinical Research & Lab Centre, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| |
Collapse
|
31
|
Chen NX, O'Neill KD, Dominguez JM, Moe SM. Regulation of reactive oxygen species in the pathogenesis of matrix vesicles induced calcification of recipient vascular smooth muscle cells. Vasc Med 2021; 26:585-594. [PMID: 34338093 DOI: 10.1177/1358863x211024721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Increased oxidative stress is associated with vascular calcification in patients with chronic kidney disease (CKD). We have previously demonstrated that cellular-derived matrix vesicles (MV), but not media-derived MV, are endocytosed in the presence of phosphorus by recipient normal rat vascular smooth muscle cells (VSMC) and induce calcification through ERK1/2 and [Ca2+]i signaling. We hypothesized that these changes were mediated by increased reactive oxygen species (ROS) production. METHODS MV were co-cultured with recipient VSMC in the presence of high phosphorus and ROS production and cell signaling assessed. RESULTS The results demonstrated MV endocytosis led to increased ROS production in recipient VSMC with no increase in mitochondrial oxygen consumption or oxidative phosphorylation (OXPHOS), indicating the ROS was not from the mitochondria. The use of inhibitors demonstrated that endocytosis of these MV by VSMC led to a signaling cascade in the cytoplasm beginning with ERK1/2 signaling, then increased [Ca2+]i and stimulation of ROS production, mediated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX)1/4. Media-derived MV did not induce this cascade, indicating endocytosis itself was not a factor. Furthermore, inhibition of either ERK1/2 activation or [Ca2+]i reduced vascular calcification. CONCLUSION We conclude that endocytosis of pro-mineralizing MV can induce a series of signaling events in normal VSMC that culminate in generation of ROS via activation of NOX1/4. Understanding these pathways will allow the development of future targeted therapeutics.
Collapse
Affiliation(s)
- Neal X Chen
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kalisha D O'Neill
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James M Dominguez
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sharon M Moe
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Roduebush Veterans Affairs Medical Center, Indianapolis, IN, USA
| |
Collapse
|
32
|
Shen A, Jiang L, Tian Y, Lu Y, Wang Z, Shen H, Song K, Feng S. Valvular Calcific Deposits and Mortality in Peritoneal Dialysis Patients: A Propensity Score-Matched Cohort Analysis. Cardiorenal Med 2021; 11:200-207. [PMID: 34320501 DOI: 10.1159/000516285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/27/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study aimed to compare mortality between peritoneal dialysis (PD) patients with and without cardiac valve calcification (CVC). METHODS Patients undergoing PD at the dialysis center of the Second Affiliated Hospital of Soochow University from January 1, 2009, to June 31, 2016, were included and followed through December 31, 2018. The inclusion criteria were (1) age ≥18 years and (2) PD vintage ≥1 month. The exclusion criteria were (1) a history of hemodialysis or renal transplantation before PD; (2) diagnosed congenital heart disease, rheumatic heart disease, or hyperthyroid heart disease; and (3) loss to follow-up. Differences in mortality rates were compared using a Fine-Gray proportional hazards model. RESULTS A total of 310 patient cases were included in this study, including 237 cases without CVC (non-CVC group). The CVC group included 59 cases with aortic valve calcification (AVC), 6 cases with mitral valve calcification (MVC), and 8 cases of AVC associated with MVC. After propensity score matching, 68 pairs were selected. The multivariate competing risk regression analysis revealed that age (hazard ratio [HR]: 1.06, 95% confidence interval [95% CI]: 1.03-1.10, p < 0.001) and CVC group (HR: 1.83, 95% CI: 1.04-3.20, p < 0.05) were independent risk factors associated with mortality. No significant difference was observed in technique survival between the 2 groups. CONCLUSION CVC is an independent risk factor for mortality in PD patients.
Collapse
Affiliation(s)
- Aiwen Shen
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, China,
| | - Linsen Jiang
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunhuan Tian
- Department of Nephrology, East District of Suzhou Municipal Hospital, Suzhou, China
| | - Ying Lu
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi Wang
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Huaying Shen
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Song
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Sheng Feng
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
33
|
Thomas DC, Thomas P, Sivan A, Unnam P, Ajayakumar A, Kumar SS, Pitchumani PK, Fatahzadeh M, Mahmud NEH. Monckeberg's Medial Sclerosis as a Cause for Headache and Facial Pain. Curr Pain Headache Rep 2021; 25:50. [PMID: 34086132 DOI: 10.1007/s11916-021-00965-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 01/02/2023]
Abstract
PURPOSE OF REVIEW Mönckeberg's medial sclerosis (MMS) is a chronic, non-inflammatory degenerative condition affecting primarily the tunica media of muscular arteries resulting in their calcification. The purpose of this comprehensive review is to describe MMS as it appears in the literature, in the context of headache and facial pain. Understanding the etiopathology, the associated conditions, and the differential diagnoses is important in managing MMS. RECENT FINDINGS Management of MMS primarily depends upon identification of its associated conditions and their treatment. Due to the rare incidence and inadequate literature on MMS presenting with headaches, the diagnosis of the pain and the entity itself is challenging. MMS is characterized by associated systemic conditions and absence of inflammatory markers. It can mimic giant cell arteritis (GCA) and other pain entities. An interdisciplinary approach involving appropriate specialties is recommended.
Collapse
Affiliation(s)
- Davis C Thomas
- Center for TMD and Orofacial Pain, Rutgers School of Dental Medicine, Newark, NJ, USA.
| | - Prisly Thomas
- Believers Church Medical College Hospital, Kerala, India
| | | | - Priyanka Unnam
- Diagnostic Sciences, Rutgers School of Dental Medicine, Newark, NJ, USA
| | | | | | | | - Mahnaz Fatahzadeh
- Division of Oral Medicine, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - Nida-E-Haque Mahmud
- Professional Program for International Dentists, University of California Los A ngeles School of Dentistry, Los Angeles, CA, USA
| |
Collapse
|
34
|
Wei X, Su Y, Li Q, Zheng Z, Hou P. Analysis of crucial genes, pathways and construction of the molecular regulatory networks in vascular smooth muscle cell calcification. Exp Ther Med 2021; 21:589. [PMID: 33850561 PMCID: PMC8027762 DOI: 10.3892/etm.2021.10021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
Vascular calcification (VC) accompanies the trans-differentiation of vascular smooth muscle cells (VSMCs) into osteo/chondrocyte-like cells and resembles physiological bone mineralization. However, the molecular mechanisms underlying VC initiation and progression have remained largely elusive. The aim of the present study was to identify the genes and pathways common to VSMC and osteoblast calcification and construct a regulatory network of non-coding RNAs and transcription factors (TFs). To this end, the Gene Expression Omnibus dataset GSE37558 including mRNA microarray data of calcifying VSMCs (CVSMCs) and calcifying osteoblasts (COs) was analyzed. The differentially expressed genes (DEGs) were screened and functionally annotated and the microRNA (miRNA/mRNA)-mRNA, TF-miRNA and long non-coding RNA (lncRNA)-TF regulatory networks were constructed. A total of 318 DEGs were identified in the CVSMCs relative to the non-calcified VSMCs, of which 43 were shared with the COs. The CVSMC-related DEGs were mainly enriched in the functional terms cell cycle, extracellular matrix (ECM), inflammation and chemotaxis-mediated signaling pathways, of which ECM was enriched by the DEGs for the COs as well. The protein-protein interaction network of CVSMCs consisted of 281 genes and 3,650 edges. There were 30 hub genes in this network, including maternal embryonic leucine zipper kinase (MELK), which potentially regulates the differentially expressed TF (DETF) forkhead box (FOX)M1 and is a potential target gene of Homo sapiens miR-485-3p and miR-181d. The TF-miRNA network included 251 TFs and 60 miRNAs, including 10 DETFs such as FOXO1 and snail family transcriptional repressor 2 (SNAI2). Furthermore, the lncRNAs H19 imprinted maternally expressed transcript (H19) and differentiation antagonizing non-protein coding RNA (DANCR) were predicted as the upstream regulators of FOXO1 and SNAI2 in the lncRNA-TF regulatory network. DANCR, MELK and FOXM1 were downregulated, and H19, FOXO1 and SNAI2 were upregulated in the CVSMCs. Taken together, the CVSMCs and COs exhibited similar molecular changes in the ECM. In addition, the MELK-FOXM1, H19/DANCR-FOXO1 and SNAI2 regulatory pathways likely mediate VSMC calcification.
Collapse
Affiliation(s)
- Xiaomin Wei
- Department of Vascular Surgery, Liuzhou Worker's Hospital, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, P.R. China
| | - Yiming Su
- Department of Vascular Surgery, Liuzhou Worker's Hospital, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, P.R. China
| | - Qiyi Li
- Department of Vascular Surgery, Liuzhou Worker's Hospital, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, P.R. China
| | - Zhiyong Zheng
- Department of Vascular Surgery, Liuzhou Worker's Hospital, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, P.R. China
| | - Peiyong Hou
- Department of Vascular Surgery, Liuzhou Worker's Hospital, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, P.R. China
| |
Collapse
|
35
|
Cai Y, Yao H, Sun Z, Wang Y, Zhao Y, Wang Z, Li L. Role of NFAT in the Progression of Diabetic Atherosclerosis. Front Cardiovasc Med 2021; 8:635172. [PMID: 33791348 PMCID: PMC8006278 DOI: 10.3389/fcvm.2021.635172] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/15/2021] [Indexed: 12/15/2022] Open
Abstract
Nuclear factor of activated T cells (NFAT) is a transcription factor with a multidirectional regulatory function, that is widely expressed in immune cells, including cells in the cardiovascular system, and non-immune cells. A large number of studies have confirmed that calcineurin/NFAT signal transduction is very important in the development of vascular system and cardiovascular system during embryonic development, and plays some role in the occurrence of vascular diseases such as atherosclerosis, vascular calcification, and hypertension. Recent in vitro and in vivo studies have shown that NFAT proteins and their activation in the nucleus and binding to DNA-related sites can easily ɨnduce the expression of downstream target genes that participate in the proliferation, migration, angiogenesis, and vascular inflammation of vascular wall related cells in various pathophysiological states. NFAT expression is regulated by various signaling pathways, including CD137-CD137L, and OX40-OX40L pathways. As a functionally diverse transcription factor, NFAT interacts with a large number of signaling molecules to modulate intracellular and extracellular signaling pathways. These NFAT-centered signaling pathways play important regulatory roles in the progression of atherosclerosis, such as in vascular smooth muscle cell phenotypic transition and migration, endothelial cell injury, macrophage-derived foam cell formation, and plaque calcification. NFAT and related signaling pathways provide new therapeutic targets for vascular diseases such as atherosclerosis. Hence, further studies of the mechanism of NFAT in the occurrence and evolution of atherosclerosis remain crucial.
Collapse
Affiliation(s)
- Yaoyao Cai
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Haipeng Yao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ying Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yunyun Zhao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
36
|
Hsu JJ, Tintut Y, Demer LL. A biomarker for vascular calcification: shedding light on an unfinished story? Cardiovasc Res 2021; 117:1809-1810. [PMID: 33751037 DOI: 10.1093/cvr/cvab071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jeffrey J Hsu
- Department of Medicine, University of California-Los Angeles, 650 Charles E. Young Dr. South, Los Angeles, CA 90095, USA.,Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA
| | - Yin Tintut
- Department of Medicine, University of California-Los Angeles, 650 Charles E. Young Dr. South, Los Angeles, CA 90095, USA.,Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA.,Department of Physiology, University of California-Los Angeles, 650 Charles E. Young Dr. South, Los Angeles, CA90095, USA.,Department of Orthopaedic Surgery, University of California-Los Angeles, 650 Charles E. Young Dr. South, Los Angeles, CA 90095, USA
| | - Linda L Demer
- Department of Medicine, University of California-Los Angeles, 650 Charles E. Young Dr. South, Los Angeles, CA 90095, USA.,Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, 11301 Wilshire Blvd., Los Angeles, CA 90073, USA.,Department of Physiology, University of California-Los Angeles, 650 Charles E. Young Dr. South, Los Angeles, CA90095, USA.,Department of Bioengineering, University of California-Los Angeles, 650 Charles E. Young Dr. South, Los Angeles, CA 90095, USA
| |
Collapse
|
37
|
Odashima K, Shimizu Y, Sano Y, Yamamoto A, Mukai T, Takada Y, Yanagisawa Y, Imai Y, Takahashi T, Kumamoto H. Osteogenic response under the periosteum by magnesium implantation in rat tibia. Dent Mater J 2021; 40:498-507. [PMID: 33642444 DOI: 10.4012/dmj.2020-011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study was designed to examine osteoconductive effects of Mg in rats tibia. The animals were sacrificed after 1, 2, and 8 weeks. The elemental analysis was performed using SEM/EDX at week 1. Following X-ray micrography at weeks 2 and 8, samples were embedded in paraffin. The expression of osteocalcin was observed by immunohistochemical staining. The element concentrations of fibrous capsules around the specimens were also measured by ICP-MS. The concentrations of Ca and P on the surface of the Mg specimen increased in SEM/EDX. The tissue specimen showed new bone formation on the bone surface near the implanted area. The concentrations of Mg, Ca, and P were high in the fibrous capsules surrounding Mg. Implantation induced differentiation of osteoblasts, and this process was considered to be associated with new bone formation. Induction of cell differentiation may be influenced by corrosion products in addition to corroding magnesium.
Collapse
Affiliation(s)
- Kenji Odashima
- Division of Oral and Maxillofacial Surgery, Department of Oral and Medicine and Surgery, Graduate School of Dentistry, Tohoku University
| | - Yoshinaka Shimizu
- Division of Oral Pathology, Department of Oral Medicine and Surgery, Graduate School of Dentistry, Tohoku University
| | - Yuya Sano
- Division of Oral Pathology, Department of Oral Medicine and Surgery, Graduate School of Dentistry, Tohoku University
| | - Akiko Yamamoto
- Biometals Group, Biomaterials Unit, Nano-life Field, International Center for Materials Nanoarchitectonics, National Institute for Materials Science
| | - Toshiji Mukai
- Department of Mechanical Engineering, Kobe University
| | - Yukyo Takada
- Division of Dental Biomaterials, Tohoku University Graduate School of Dentistry, Tohoku University
| | - Yuta Yanagisawa
- Division of Oral and Maxillofacial Surgery, Department of Oral and Medicine and Surgery, Graduate School of Dentistry, Tohoku University
| | - Yoshimichi Imai
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine
| | - Tetsu Takahashi
- Division of Oral and Maxillofacial Surgery, Department of Oral and Medicine and Surgery, Graduate School of Dentistry, Tohoku University
| | - Hiroyuki Kumamoto
- Division of Oral Pathology, Department of Oral Medicine and Surgery, Graduate School of Dentistry, Tohoku University
| |
Collapse
|
38
|
Lee DO, Kim JH, Kang HW, Kim DY, Lee W, Cho TJ, Lee DY. Calcifying characteristics of peripheral vascular smooth muscle cells of chronic kidney disease patients with critical limb ischemia. Vasc Med 2021; 26:139-146. [PMID: 33591894 DOI: 10.1177/1358863x20984525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The role of vascular smooth muscle cells (VSMCs) in vascular calcification, which is related to chronic kidney disease (CKD), has been studied in greater detail in the major arteries relative to the peripheral arteries. We compared the calcifying characteristics of peripheral VSMCs relative to non-pathologic major VSMCs in patients with severe peripheral artery disease (PAD). We isolated peripheral VSMCs from the posterior tibial artery of 10 patients with CKD who underwent below-knee amputation for critical limb ischemia (CLI). Using normal human aortic VSMCs as a control group, we cultured the cells in normal and high phosphate media for 10 days, and subsequently tested by immunofluorescence staining. We compared the calcification levels between the two groups using various assays, tests for cell viability, and scanning electron microscopy. As a result, calcification of pathologic peripheral VSMCs increased significantly with time (p = 0.028) and was significantly higher than that in human aortic VSMCs in calcium assays (p = 0.043). Dead cells in the pathologic VSMC group were more distinct in high phosphate media than in human aortic VSMCs. In conclusion, VSMCs from the peripheral artery of patients with severe CKD and CLI who underwent amputation surgery showed marked calcifying characteristics compared to normal human aortic VSMCs.
Collapse
Affiliation(s)
- Dong-Oh Lee
- Department of Orthopedic Surgery, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Ji Hye Kim
- Department of Orthopedic Surgery, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Ho Won Kang
- Department of Orthopedic Surgery, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Dae Yoo Kim
- Department of Orthopedic Surgery, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Wonik Lee
- Department of Orthopedic Surgery, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Tae-Joon Cho
- Department of Orthopedic Surgery, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Dong Yeon Lee
- Department of Orthopedic Surgery, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| |
Collapse
|
39
|
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: 3.8] [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
|
40
|
Abstract
Phosphorus plays a vital role in diverse biological processes including intracellular signaling, membrane integrity, and skeletal biomineralization; therefore, the regulation of phosphorus homeostasis is essential to the well-being of the organism. Cells and whole organisms respond to changes in inorganic phosphorus (Pi) concentrations in their environment by adjusting Pi uptake and altering biochemical processes in cells (local effects) and distant organs (endocrine effects). Unicellular organisms, such as bacteria and yeast, express specific Pi-binding proteins on the plasma membrane that respond to changes in ambient Pi availability and transduce intracellular signals that regulate the expression of genes involved in cellular Pi uptake. Multicellular organisms, including humans, respond at a cellular level to adapt to changes in extracellular Pi concentrations and also have endocrine pathways which integrate signals from various organs (e.g., intestine, kidneys, parathyroid glands, bone) to regulate serum Pi concentrations and whole-body phosphorus balance. In mammals, alterations in the concentrations of extracellular Pi modulate type III sodium-phosphate cotransporter activity on the plasma membrane, and trigger changes in cellular function. In addition, elevated extracellular Pi induces activation of fibroblast growth factor receptor, Raf/mitogen-activated protein kinase/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) and Akt pathways, which modulate gene expression in various mammalian cell types. Excessive Pi exposure, especially in patients with chronic kidney disease, leads to endothelial dysfunction, accelerated vascular calcification, and impaired insulin secretion.
Collapse
Affiliation(s)
- Kittrawee Kritmetapak
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Division of Nephrology and Hypertension, Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55902, USA
| | - Rajiv Kumar
- Division of Nephrology and Hypertension, Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55902, USA.
- Nephrology Research, Medical Sciences 1-120, 200 First Street Southwest, Rochester, MN, 55902, USA.
| |
Collapse
|
41
|
Chao CT, Lin SH. Uremic Vascular Calcification: The Pathogenic Roles and Gastrointestinal Decontamination of Uremic Toxins. Toxins (Basel) 2020; 12:toxins12120812. [PMID: 33371477 PMCID: PMC7767516 DOI: 10.3390/toxins12120812] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 12/16/2022] Open
Abstract
Uremic vascular calcification (VC) commonly occurs during advanced chronic kidney disease (CKD) and significantly increases cardiovascular morbidity and mortality. Uremic toxins are integral within VC pathogenesis, as they exhibit adverse vascular influences ranging from atherosclerosis, vascular inflammation, to VC. Experimental removal of these toxins, including small molecular (phosphate, trimethylamine-N-oxide), large molecular (fibroblast growth factor-23, cytokines), and protein-bound ones (indoxyl sulfate, p-cresyl sulfate), ameliorates VC. As most uremic toxins share a gut origin, interventions through gastrointestinal tract are expected to demonstrate particular efficacy. The “gastrointestinal decontamination” through the removal of toxin in situ or impediment of toxin absorption within the gastrointestinal tract is a practical and potential strategy to reduce uremic toxins. First and foremost, the modulation of gut microbiota through optimizing dietary composition, the use of prebiotics or probiotics, can be implemented. Other promising strategies such as reducing calcium load, minimizing intestinal phosphate absorption through the optimization of phosphate binders and the inhibition of gut luminal phosphate transporters, the administration of magnesium, and the use of oral toxin adsorbent for protein-bound uremic toxins may potentially counteract uremic VC. Novel agents such as tenapanor have been actively tested in clinical trials for their potential vascular benefits. Further advanced studies are still warranted to validate the beneficial effects of gastrointestinal decontamination in the retardation and treatment of uremic VC.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan;
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Shih-Hua Lin
- Department of Internal Medicine, Tri-Service General Hospital and National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence:
| |
Collapse
|
42
|
Li Y, Sun W, Saaoud F, Wang Y, Wang Q, Hodge J, Hui Y, Yin S, Lessner SM, Kong X, Fan D. MiR155 modulates vascular calcification by regulating Akt-FOXO3a signalling and apoptosis in vascular smooth muscle cells. J Cell Mol Med 2020; 25:535-548. [PMID: 33210462 PMCID: PMC7810936 DOI: 10.1111/jcmm.16107] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/13/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022] Open
Abstract
microRNA‐155 (miR155) is pro‐atherogenic; however, its role in vascular calcification is unknown. In this study, we aim to examine whether miR155 regulates vascular calcification and to understand the underlying mechanism. Quantitative real‐time PCR showed that miR155 is highly expressed in human calcific carotid tissue and positively correlated with the expression of osteogenic genes. Wound‐healing assay and TUNEL staining showed deletion of miR155 inhibited vascular smooth muscle cell (VSMC) migration and apoptosis. miR155 deficiency attenuated calcification of cultured mouse VSMCs and aortic rings induced by calcification medium, whereas miR155 overexpression promoted VSMC calcification. Compared with wild‐type mice, miR155−/− mice showed significant resistance to vitamin D3 induced vascular calcification. Protein analysis showed that miR155 deficiency alleviated the reduction of Rictor, increased phosphorylation of Akt at S473 and accelerated phosphorylation and degradation of FOXO3a in cultured VSMCs and in the aortas of vitamin D3‐treated mice. A PI3K inhibitor that suppresses Akt phosphorylation increased, whereas a pan‐caspase inhibitor that suppresses apoptosis reduced VSMC calcification; and both inhibitors diminished the protective effects of miR155 deficiency on VSMC calcification. In conclusion, miR155 deficiency attenuates vascular calcification by increasing Akt phosphorylation and FOXO3a degradation, and thus reducing VSMC apoptosis induced by calcification medium.
Collapse
Affiliation(s)
- Yong Li
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Wei Sun
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.,Department of Cardiology and Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fatma Saaoud
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Quanyi Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Johnie Hodge
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Yvonne Hui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Sophia Yin
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Susan M Lessner
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Xiangqing Kong
- Department of Cardiology and Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
43
|
Hill Gallant KM, Stremke ER, Trevino LL, Moorthi RN, Doshi S, Wastney ME, Hisada N, Sato J, Ogita Y, Fujii N, Matsuda Y, Kake T, Moe SM. EOS789, a broad-spectrum inhibitor of phosphate transport, is safe with an indication of efficacy in a phase 1b randomized crossover trial in hemodialysis patients. Kidney Int 2020; 99:1225-1233. [PMID: 33137340 DOI: 10.1016/j.kint.2020.09.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022]
Abstract
The treatment of hyperphosphatemia remains challenging in patients receiving hemodialysis. This phase 1b study assessed safety and efficacy of EOS789, a novel pan-inhibitor of phosphate transport (NaPi-2b, PiT-1, PiT-2) on intestinal phosphate absorption in patients receiving intermittent hemodialysis therapy. Two cross-over, randomized order studies of identical design (ten patients each) compared daily EOS789 50 mg to placebo with meals and daily EOS789 100 mg vs EOS789 100 mg plus 1600 mg sevelamer with meals. Patients ate a controlled diet of 900 mg phosphate daily for two weeks and began EOS789 on day four. On day ten, a phosphate absorption testing protocol was performed during the intradialytic period. Intestinal fractional phosphate absorption was determined by kinetic modeling of serum data following oral and intravenous doses of 33Phosphate (33P). The results demonstrated no study drug related serious adverse events. Fractional phosphate absorption was 0.53 (95% confidence interval: 0.39,0.67) for placebo vs. 0.49 (0.35,0.63) for 50 mg EOS789; and 0.40 (0.29,0.50) for 100 mg EOS789 vs. 0.36 (0.26,0.47) for 100 mg EOS789 plus 1600 mg sevelamer (all not significantly different). The fractional phosphate absorption trended lower in six patients who completed both studies with EOS789 100 mg compared with placebo. Thus, in this phase 1b study, EOS789 was safe and well tolerated. Importantly, the use of 33P as a sensitive and direct measure of intestinal phosphate absorption allows specific testing of drug efficacy. The effectiveness of EOS789 needs to be evaluated in future phase 2 and phase 3 studies.
Collapse
Affiliation(s)
- Kathleen M Hill Gallant
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA; Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Elizabeth R Stremke
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| | - Laurie L Trevino
- Clinical Translational Sciences Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ranjani N Moorthi
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Simit Doshi
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Meryl E Wastney
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA; Metabolic Modeling Services, West Lafayette, Indiana, USA
| | | | | | | | | | | | | | - Sharon M Moe
- Clinical Translational Sciences Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA; Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
44
|
Piri R, Gerke O, Høilund-Carlsen PF. Molecular imaging of carotid artery atherosclerosis with PET: a systematic review. Eur J Nucl Med Mol Imaging 2020; 47:2016-2025. [PMID: 31786626 DOI: 10.1007/s00259-019-04622-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE To conduct a systematic review of articles on PET imaging of carotid atherosclerosis with emphasis on clinical usefulness and comparison with other imaging modalities. METHODS Research articles reporting carotid artery PET imaging with different radiotracers until 30 November 2018 were systematically searched for in Medline/PubMed, Scopus, Embase, Google Scholar, and Cochrane Library. Duplicates were removed, and editorials, case studies, and investigations on feasibility or reproducibility of PET imaging and of patients with end-stage diseases or immunosuppressive medications were omitted. After quality assessment of included articles using Joanna Briggs Institute checklists, all eligible articles were reviewed. RESULTS Of 1718 primary hits, 53 studies comprising 4472 patients, aged 47-91 years (78.8% males), were included and grouped under the following headlines: diagnostic performance, risk factors, laboratory findings, imaging modalities, and treatment. 18F-fluorodeoxyglucose (FDG) (49/53) and 18F-sodium fluoride (NaF) (5/53) were the most utilized tracers to visualize carotid wall inflammation and microcalcification, respectively. Higher carotid FDG uptake was demonstrated in patients with than without symptomatic carotid atherosclerosis. Normal carotid arteries presented with the lowest FDG uptake. In symptomatic atherosclerosis, carotid arteries ipsilateral to a cerebrovascular event had higher FDG uptake than the contralateral carotid artery. FDG uptake was significantly associated with age, male gender, and body mass index in healthy individuals, and in addition with arterial hypertension, hypercholesterolemia, and diabetes mellitus in patients. Histological assessment indicated a strong correlation between microcalcification and NaF uptake in symptomatic patients. Histological evidence of calcification correlated inversely with FDG uptake, which was associated with increased macrophage and CD68 count, both accounting for increased local inflammatory response. CONCLUSION FDG-PET visualizes the inflammatory part of carotid atherosclerosis enabling risk stratification to a certain degree, whereas NaF-PET seems to indicate long-term consequences of ongoing inflammation by demonstrating microcalcification allowing discrimination of atherosclerotic from normal arteries and suggesting clinically significant carotid atherosclerosis.
Collapse
Affiliation(s)
- Reza Piri
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark.
- Research Unit of Clinical Physiology and Nuclear Medicine, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Oke Gerke
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Research Unit of Clinical Physiology and Nuclear Medicine, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Research Unit of Clinical Physiology and Nuclear Medicine, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
45
|
Li C, Zhang S, Chen X, Ji J, Yang W, Gui T, Gai Z, Li Y. Farnesoid X receptor activation inhibits TGFBR1/TAK1-mediated vascular inflammation and calcification via miR-135a-5p. Commun Biol 2020; 3:327. [PMID: 32581266 PMCID: PMC7314757 DOI: 10.1038/s42003-020-1058-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 06/08/2020] [Indexed: 01/06/2023] Open
Abstract
Chronic inflammation plays a crucial role in vascular calcification. However, only a few studies have revealed the mechanisms underlying the development of inflammation under high-phosphate conditions in chronic kidney disease (CKD) patients. Here, we show that inflammation resulting from the activation of the TGFBR1/TAK1 pathway is involved in calcification in CKD rats or osteogenic medium-cultured human aortic smooth muscle cells (HASMCs). Moreover, miR-135a-5p is demonstrated to be a key regulator of the TGFBR1/TAK1 pathway, which has been reported to be decreased in CKD rats. We further reveal that farnesoid X receptor (FXR) activation increases miR-135a-5p expression, thereby inhibiting the activation of the TGFBR1/TAK1 pathway, ultimately resulting in the attenuation of vascular inflammation and calcification in CKD rats. Our findings provide advanced insights into the mechanisms underlying the development of inflammation in vascular calcification, and evidence that FXR activation could serve as a therapeutic strategy for retarding vascular calcification in CKD patients.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Calcinosis/genetics
- Calcinosis/metabolism
- Cells, Cultured
- Female
- Humans
- MAP Kinase Kinase Kinases/genetics
- MAP Kinase Kinase Kinases/metabolism
- Male
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/cytology
- Osteogenesis
- Rats, Wistar
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/pathology
- Vasculitis/genetics
- Vasculitis/metabolism
- Vasculitis/pathology
Collapse
Affiliation(s)
- Chao Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, 8032, Switzerland
| | - Shijun Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaoqing Chen
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jingkang Ji
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenqing Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Ting Gui
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhibo Gai
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, 8032, Switzerland.
| | - Yunlun Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, China.
| |
Collapse
|
46
|
Sanchis P, Rivera R, Fortuny R, Río C, Mas-Gelabert M, Gonzalez-Freire M, Grases F, Masmiquel L. Role of Advanced Glycation End Products on Aortic Calcification in Patients with Type 2 Diabetes Mellitus. J Clin Med 2020; 9:jcm9061751. [PMID: 32516928 PMCID: PMC7356630 DOI: 10.3390/jcm9061751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to evaluate the relationship between serum levels of advanced glycation end products (AGEs) and abdominal aortic calcification (AAC) in patients with type 2 diabetes mellitus (DM2). This was a prospective cross-sectional study. One-hundred and four consecutive patients with DM2 were given lateral lumbar X-rays in order to quantify abdominal aortic calcification (AAC). Circulating levels of AGEs and classical cardiovascular risk factors were determined. Clinical history was also registered. Patients with higher AGEs values had higher grades of aortic calcification and higher numbers of diabetic-related complications. Multivariate logistic regression analysis showed that being older, male and having high levels of AGEs and triglycerides were the independent risk factors associated to moderate-severe AAC when compared to no-mild AAC. Our results suggest that AGEs plays a role in the pathogenesis of aortic calcifications. In addition, the measurement of AGEs levels may be useful for assessing the severity of AAC in the setting of diabetic complications.
Collapse
Affiliation(s)
- Pilar Sanchis
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands [IUNICS-IdISBa], 07198 Palma of Mallorca, Spain; (R.R.); (R.F.); (M.G.-F.)
- Laboratory of Renal Lithiasis Research, Deptartment of Chemistry, University of Balearic Islands, Health Research Institute of the Balearic Islands [IUNICS-IdISBa], 07122 Palma of Mallorca, Spain;
- The Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, 28046 Madrid, Spain
- Correspondence: (P.S.); (L.M.)
| | - Rosmeri Rivera
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands [IUNICS-IdISBa], 07198 Palma of Mallorca, Spain; (R.R.); (R.F.); (M.G.-F.)
| | - Regina Fortuny
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands [IUNICS-IdISBa], 07198 Palma of Mallorca, Spain; (R.R.); (R.F.); (M.G.-F.)
- Laboratory Department, Son Llàtzer University Hospital, 07198 Palma of Mallorca, Spain
| | - Carlos Río
- Proteomics department, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma of Mallorca, Spain;
| | - Miguel Mas-Gelabert
- Radiology Department, Son Llàtzer University Hospital, 07198 Palma of Mallorca, Spain;
| | - Marta Gonzalez-Freire
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands [IUNICS-IdISBa], 07198 Palma of Mallorca, Spain; (R.R.); (R.F.); (M.G.-F.)
| | - Felix Grases
- Laboratory of Renal Lithiasis Research, Deptartment of Chemistry, University of Balearic Islands, Health Research Institute of the Balearic Islands [IUNICS-IdISBa], 07122 Palma of Mallorca, Spain;
- The Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, 28046 Madrid, Spain
| | - Luis Masmiquel
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands [IUNICS-IdISBa], 07198 Palma of Mallorca, Spain; (R.R.); (R.F.); (M.G.-F.)
- Correspondence: (P.S.); (L.M.)
| |
Collapse
|
47
|
Li Y, Sun Z, Zhang L, Yan J, Shao C, Jing L, Li L, Wang Z. Role of Macrophages in the Progression and Regression of Vascular Calcification. Front Pharmacol 2020; 11:661. [PMID: 32457633 PMCID: PMC7227444 DOI: 10.3389/fphar.2020.00661] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification is an abnormal cell-mediated process in which bone-specific hydroxyapatite crystals are actively deposited on the blood vessel wall and is a significant pathological basis for the increased incidence and mortality of adverse cardiovascular events. Macrophages play an important regulatory role in the occurrence, development, and regression of vascular calcification. After the tissue microenvironment changes, macrophages subsequently change their polarity and phenotype or secrete functional substances as an adaptive response. As research on macrophages continue to move into this field, we gain a new understanding of the mechanism of the formation and regression of vascular calcification, which might offer valuable new intervention targets for the prevention and inhibition of vascular calcification. This review summarizes a wealth of research in this field and explores the roles of macrophages in the development process of vascular calcification.
Collapse
Affiliation(s)
- Yalan Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jinchuan Yan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lele Jing
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
48
|
Bolean M, Izzi B, van Kerckhoven S, Bottini M, Ramos AP, Millán JL, Hoylaerts MF, Ciancaglini P. Matrix vesicle biomimetics harboring Annexin A5 and alkaline phosphatase bind to the native collagen matrix produced by mineralizing vascular smooth muscle cells. Biochim Biophys Acta Gen Subj 2020; 1864:129629. [PMID: 32360152 DOI: 10.1016/j.bbagen.2020.129629] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/13/2020] [Accepted: 04/28/2020] [Indexed: 10/24/2022]
Abstract
BACKGOUND Vascular smooth muscle cells (VSMCs) transdifferentiated ectopically trigger vascular calcifications, contributing to clinical cardiovascular disease in the aging population. AnxA5 and TNAP play a crucial role in (patho)physiological mineralization. METHODS We performed affinity studies between DPPC and 9:1 DPPC:DPPS-proteoliposomes carrying AnxA5 and/or TNAP and different types of collagen matrix: type I, II, I + III and native collagenous extracellular matrix (ECM) produced from VSMCs with or without differentiation, to simulate ectopic calcification conditions. RESULTS AnxA5-proteoliposomes had the highest affinity for collagens, specially for type II. TNAP-proteoliposomes bound poorly and the simultaneous presence of TNAP in the AnxA5-proteoliposomes disturbed interactions between AnxA5 and collagen. DPPC AnxA5-proteoliposomes affinities for ECM from transdifferentiating cells went up 2-fold compared to that from native VSMCs. The affinities of DPPC:DPPS-proteoliposomes were high for ECM from VSMCs with or without differentiation, underscoring a synergistic effect between AnxA5 and DPPS. Co-localization studies uncovered binding of proteoliposomes harboring AnxA5 or TNAP+AnxA5 to various regions of the ECM, not limited to type II collagen. CONCLUSION AnxA5-proteoliposomes showed the highest affinities for type II collagen, deposited during chondrocyte mineralization in joint cartilage. TNAP in the lipid/protein microenvironment disturbs interactions between AnxA5 and collagen. These findings support the hypothesis that TNAP is cleaved from the MVs membrane just before ECM binding, such facilitating MV anchoring to ECM via AnxA5 interaction. GENERAL SIGNIFICANCE Proteoliposomes as MV biomimetics are useful in the understanding of mechanisms that regulate the mineralization process and may be essential for the development of novel therapeutic strategies to prevent or inhibit ectopic mineralization.
Collapse
Affiliation(s)
- Maytê Bolean
- Department of Chemistry, FFCLRP-USP, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Benedetta Izzi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, IS, Italy
| | | | - Massimo Bottini
- University of Rome Tor Vergata, Department of Experimental Medicine and Surgery, Roma, Italy; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ana Paula Ramos
- Department of Chemistry, FFCLRP-USP, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - José Luis Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Marc F Hoylaerts
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Pietro Ciancaglini
- Department of Chemistry, FFCLRP-USP, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
49
|
Chen NX, Srinivasan S, O’Neill K, Nickolas TL, Wallace JM, Allen MR, Metzger CE, Creecy A, Avin KG, Moe SM. Effect of Advanced Glycation End-Products (AGE) Lowering Drug ALT-711 on Biochemical, Vascular, and Bone Parameters in a Rat Model of CKD-MBD. J Bone Miner Res 2020; 35:608-617. [PMID: 31743501 PMCID: PMC9030558 DOI: 10.1002/jbmr.3925] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 11/04/2019] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease-mineral bone disorder (CKD-MBD) is a systemic disorder that affects blood measures of bone and mineral homeostasis, vascular calcification, and bone. We hypothesized that the accumulation of advanced glycation end-products (AGEs) in CKD may be responsible for the vascular and bone pathologies via alteration of collagen. We treated a naturally occurring model of CKD-MBD, the Cy/+ rat, with a normal and high dose of the AGE crosslink breaker alagebrium (ALT-711), or with calcium in the drinking water to mimic calcium phosphate binders for 10 weeks. These animals were compared to normal (NL) untreated animals. The results showed that CKD animals, compared to normal animals, had elevated blood urea nitrogen (BUN), PTH, FGF23 and phosphorus. Treatment with ALT-711 had no effect on kidney function or PTH, but 3 mg/kg lowered FGF23 whereas calcium lowered PTH. Vascular calcification of the aorta assessed biochemically was increased in CKD animals compared to NL, and decreased by the normal, but not high dose of ALT-711, with parallel decreases in left ventricular hypertrophy. ALT-711 (3 mg/kg) did not alter aorta AGE content, but reduced aorta expression of receptor for advanced glycation end products (RAGE) and NADPH oxidase 2 (NOX2), suggesting effects related to decreased oxidative stress at the cellular level. The elevated total bone AGE was decreased by 3 mg/kg ALT-711 and both bone AGE and cortical porosity were decreased by calcium treatment, but only calcium improved bone properties. In summary, treatment of CKD-MBD with an AGE breaker ALT-711, decreased FGF23, reduced aorta calcification, and reduced total bone AGE without improvement of bone mechanics. These results suggest little effect of ALT-711 on collagen, but potential cellular effects. The data also highlights the need to better measure specific types of AGE proteins at the tissue level in order to fully elucidate the impact of AGEs on CKD-MBD. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Neal X Chen
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shruthi Srinivasan
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kalisha O’Neill
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas L Nickolas
- Division of Nephrology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Matthew R Allen
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Corinne E Metzger
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Amy Creecy
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Keith G Avin
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN, USA
| | - Sharon M Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| |
Collapse
|
50
|
Wen M, Zhi D, Wang L, Cui C, Huang Z, Zhao Y, Wang K, Kong D, Yuan X. Local Delivery of Dual MicroRNAs in Trilayered Electrospun Grafts for Vascular Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:6863-6875. [PMID: 31958006 DOI: 10.1021/acsami.9b19452] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Globally growing problems related to cardiovascular diseases lead to a considerable need for synthetic vascular grafts. For small-caliber vascular prosthesis, it remains essential to fulfill rapid endothelialization, inhibit intimal hyperplasia, and prevent calcification for keeping patency. To modulate vascular regeneration, herein, we developed a bioactive trilayered tissue-engineered vascular graft encapsulating both microRNA-126 and microRNA-145 in the fibrous inner and middle layers, respectively. In vitro cell activities demonstrated that the trilayered electrospun membranes had significant biological advantages in enhanced growth and intracellular nitric oxide production of vascular endothelial cells, modulation of phenotypes of vascular smooth muscle cells (SMCs), and restraint of calcium deposition through fast-releasing microRNA-126 and slow-releasing microRNA-145. Histological and immunofluorescent analyses of in vivo implantation in a rat abdominal aorta interposition model suggested that the dual-microRNA-loading trilayered electrospun graft exerted a positive effect on accelerating endothelialization, improving contractile SMC regeneration, and promoting normal extracellular matrix formation. Meanwhile, the local bioactivity of microRNA-126 and microRNA-145 in the trilayered vascular graft could regulate inflammation and depress calcification possibly by facilitating transformation of macrophages into the anti-inflammatory M2 phenotype. These findings indicated that the trilayered electrospun graft by local delivery of dual microRNAs could be possibly used as a bioactive substitute for replacement of artificial small-caliber blood vessels.
Collapse
Affiliation(s)
- Meiling Wen
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Dengke Zhi
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Lina Wang
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Ce Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Ziqi Huang
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Yunhui Zhao
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Kai Wang
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Deling Kong
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Xiaoyan Yuan
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| |
Collapse
|