1
|
Fu C, Liu Y, Yang H, Liang Q, Liu W, Guo W. Construction of a miR-15a-based risk prediction model for vascular calcification detection in patients undergoing hemodialysis. Ren Fail 2024; 46:2313175. [PMID: 38419564 PMCID: PMC10906117 DOI: 10.1080/0886022x.2024.2313175] [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: 07/31/2023] [Accepted: 01/27/2024] [Indexed: 03/02/2024] Open
Abstract
Vascular calcification (VC) is highly prevalent in patients undergoing hemodialysis, and is a significant contributor to the mortality rate. Therefore, biomarkers that can accurately predict the onset of VC are urgently required. Our study aimed to investigate serum miR-15a levels in relation to VC and to develop a predictive model for VC in patients undergoing hemodialysis at the Beijing Friendship Hospital hemodialysis center between 1 January 2019 and 31 December 2020. The patients were categorized into two groups: VC and non-VC. Logistic regression (LR) models were used to examine the risk factors associated with VC. Additionally, we developed an miR-15a-based nomogram based on the results of the multivariate LR analysis. A total of 138 patients under hemodialysis were investigated (age: 58.41 ± 13.22 years; 54 males). VC occurred in 79 (57.2%) patients. Multivariate LR analysis indicated that serum miR-15a, age, and WBC count were independent risk factors for VC. A miR-15a-based nomogram was developed by incorporating the following five predictors: age, dialysis vintage, predialysis nitrogen, WBC count, and miR-15a. The receiver operating characteristic (ROC) curve had an area under the curve of 0.921, diagnostic threshold of 0.396, sensitivity of 0.722, and specificity of 0.932, indicating that this model had good discrimination. This study concluded that serum miR-15a levels, age, and white blood cell (WBC) count are independent risk factors for VC. A nomogram constructed by integrating these risk factors can be used to predict the risk of VC in patients undergoing hemodialysis.
Collapse
Affiliation(s)
- Chen Fu
- Department of Nephrology, Faculty of Kidney Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Yingjie Liu
- Department of Nephrology, Faculty of Kidney Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Huayu Yang
- Division of Geriatrics, Medical and Health Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Qiaojing Liang
- Division of Geriatrics, Medical and Health Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Wenhu Liu
- Department of Nephrology, Faculty of Kidney Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Weikang Guo
- Department of Nephrology, Faculty of Kidney Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| |
Collapse
|
2
|
Xu T, Li Y, Cheng M, Jin J, Zhang S, Bai Y, Xu J. Construction of competitive endogenous RNA network and identification of potential regulatory axis in vascular calcification. FASEB J 2024; 38:e70114. [PMID: 39432302 DOI: 10.1096/fj.202400973rr] [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: 04/28/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Competitive endogenous RNAs (ceRNA) theory has been proved in numerous biological processes. Nevertheless, there is a lack of research applying the ceRNA theory to the study of vascular calcification (VC) in chronic kidney diseases (CKD). In the present study, a ceRNA network was constructed after conducting transcriptome sequencing of differentially expressed genes, followed by experimental validation to identify a new target for the diagnosis and treatment of vascular calcification. Total RNA was extracted from β-glycerophosphate (β-GP) cultured vascular smooth muscle cells (VSMCs) on Day 7. Illumina HiSeq platform was utilized to build sequencing libraries. GO and KEGG analysis was conducted to identify the function of the differentially expressed genes. Protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database. A ceRNA network was established based on TargetScan, miRDB, miRWALK, and miRanda database. Western blot and qRT-PCR were used to explore the expression level of protein and RNA, respectively. The direct binding sites were confirmed by dual-luciferase reporter assay. In total, 647 differentially expressed lncRNAs and 289 differentially expressed mRNAs were identified (|log2FC| ≥ 1, p < .05). The function of differentially expressed mRNAs was mainly enriched in negative regulation of osteoblast differentiation, regulation of RNA metabolic process, and other typical pathways. The ceRNA network was generated with a total of 107 interaction pairs. The lncRNA Prrc2c/miR-145-5p/Smad3 axis was considered a potential regulatory pathway within the ceRNA network. The regulatory relationship and targets of this ceRNA axis were validated via in vitro experiments. For the first time, we found that lncRNA Prrc2c was highly expressed and promoted calcification of VSMCs. Luciferase reporter assay showed that lncRNA Prrc2c could bind miR-145-5p at site 1755-1761. Similarly, luciferase reporter assay showed that miR-145-5p inhibited Smad3 expression by binding to its 3'UTR. Our findings provide a comprehensive examination of the ceRNA networks in vascular smooth muscle cells (VSMCs) treated with high phosphorate. Specifically, we have identified the role of lncRNA Prrc2c in promoting VSMC calcification through the miR-145-5p/Smad3 axis.
Collapse
MESH Headings
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Gene Regulatory Networks
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Humans
- Myocytes, Smooth Muscle/metabolism
- Protein Interaction Maps
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Smad3 Protein/metabolism
- Smad3 Protein/genetics
- Cells, Cultured
- Gene Expression Regulation
- Transcriptome
- Glycerophosphates/metabolism
- RNA, Competitive Endogenous
Collapse
Affiliation(s)
- Tongxin Xu
- Department of CT&MRI, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yuzhe Li
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, P.R. China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, P.R. China
| | - Meijuan Cheng
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, P.R. China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, P.R. China
| | - Jingjing Jin
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, P.R. China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, P.R. China
| | - Shenglei Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, P.R. China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, P.R. China
| | - Yaling Bai
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, P.R. China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, P.R. China
| | - Jinsheng Xu
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, P.R. China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, P.R. China
| |
Collapse
|
3
|
Wang Z, Gui Z, Zhang L, Wang Z. Advances in the mechanisms of vascular calcification in chronic kidney disease. J Cell Physiol 2024:e31464. [PMID: 39392232 DOI: 10.1002/jcp.31464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Vascular calcification (VC) is common in patients with advanced chronic kidney disease (CKD).A series of factors, such as calcium and phosphorus metabolism disorders, uremic toxin accumulation, inflammation and oxidative stress and cellular senescence, cause osteoblast-like differentiation of vascular smooth muscle cells, secretion of extracellular vesicles, and imbalance of calcium regulatory factors, which together promote the development of VC in CKD. Recent advances in epigenetics have provided better tools for the investigation of VC etiology and new approaches for finding more accurate biomarkers. These advances have not only deepened our understanding of the pathophysiological mechanisms of VC in CKD, but also provided valuable clues for the optimization of clinical predictors and the exploration of potential therapeutic targets. The aim of this article is to provide a comprehensive overview of the pathogenesis of CKD VC, especially the new advances made in recent years, including the various key factors mentioned above. Through the comprehensive analysis, we expect to provide a solid theoretical foundation and research direction for future studies targeting the specific mechanisms of CKD VC, the establishment of clinical predictive indicators and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Zebin Gui
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Lirong Zhang
- Department of Radiology, Affliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| |
Collapse
|
4
|
Fernández-Villabrille S, Martín-Carro B, Martín-Vírgala J, Rodríguez-Santamaria MDM, Baena-Huerta F, Muñoz-Castañeda JR, Fernández-Martín JL, Alonso-Montes C, Naves-Díaz M, Carrillo-López N, Panizo S. Novel Biomarkers of Bone Metabolism. Nutrients 2024; 16:605. [PMID: 38474734 DOI: 10.3390/nu16050605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Bone represents a metabolically active tissue subject to continuous remodeling orchestrated by the dynamic interplay between osteoblasts and osteoclasts. These cellular processes are modulated by a complex interplay of biochemical and mechanical factors, which are instrumental in assessing bone remodeling. This comprehensive evaluation aids in detecting disorders arising from imbalances between bone formation and reabsorption. Osteoporosis, characterized by a reduction in bone mass and strength leading to heightened bone fragility and susceptibility to fractures, is one of the more prevalent chronic diseases. Some epidemiological studies, especially in patients with chronic kidney disease (CKD), have identified an association between osteoporosis and vascular calcification. Notably, low bone mineral density has been linked to an increased incidence of aortic calcification, with shared molecules, mechanisms, and pathways between the two processes. Certain molecules emerging from these shared pathways can serve as biomarkers for bone and mineral metabolism. Detecting and evaluating these alterations early is crucial, requiring the identification of biomarkers that are reliable for early intervention. While traditional biomarkers for bone remodeling and vascular calcification exist, they suffer from limitations such as low specificity, low sensitivity, and conflicting results across studies. In response, efforts are underway to explore new, more specific biomarkers that can detect alterations at earlier stages. The aim of this review is to comprehensively examine some of the emerging biomarkers in mineral metabolism and their correlation with bone mineral density, fracture risk, and vascular calcification as well as their potential use in clinical practice.
Collapse
Affiliation(s)
- Sara Fernández-Villabrille
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Beatriz Martín-Carro
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Julia Martín-Vírgala
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | | | - Francisco Baena-Huerta
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Juan Rafael Muñoz-Castañeda
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Nephrology Service, Reina Sofia University Hospital, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Córdoba, 14004 Córdoba, Spain
| | - José Luis Fernández-Martín
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Cristina Alonso-Montes
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Manuel Naves-Díaz
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Natalia Carrillo-López
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Sara Panizo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| |
Collapse
|
5
|
Wu KL, Chen CL, Thi Nguyen MH, Tsai JC, Wang SC, Chiang WF, Hsiao PJ, Chan JS, Hou JJ, Ma N. MicroRNA regulators of vascular pathophysiology in chronic kidney disease. Clin Chim Acta 2023; 551:117610. [PMID: 37863246 DOI: 10.1016/j.cca.2023.117610] [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: 08/19/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Coronary artery disease (CAD) is a severe comorbidity in chronic kidney disease (CKD) due to heavy calcification in the medial layer and inflamed plaques. Chronic inflammation, endothelial dysfunction and vascular calcification are major contributors that lead to artherosclerosis in CKD. The lack of specific symptoms and signs of CAD and decreased accuracy of noninvasive diagnostic tools result in delayed diagnosis leading to increased mortality. MicroRNAs (miRNAs) are post-transcriptional regulators present in various biofluids throughout the body. In the circulation, miRNAs have been reported to be encapsulated in extracellular vesicles and serve as stable messengers for crosstalk among cells. miRNAs are involved in pathophysiologic mechanisms including CAD and can potentially be extended from basic research to clinical translational practice.
Collapse
Affiliation(s)
- Kun-Lin Wu
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Lung Chen
- Division of Nephrology, Department of Medicine, Landseed International Hospital, Taoyuan, Taiwan
| | - Mai-Huong Thi Nguyen
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Jen-Chieh Tsai
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan; Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Sun-Chong Wang
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Wen-Fang Chiang
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Jen Hsiao
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jenq-Shyong Chan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ju Jung Hou
- Kaohsiung Medical University Hospital, Department of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Nianhan Ma
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan.
| |
Collapse
|
6
|
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: 2.0] [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
|
7
|
Yu F, Duan Y, Liu C, Huang H, Xiao X, He Z. Extracellular vesicles in atherosclerosis and vascular calcification: the versatile non-coding RNAs from endothelial cells and vascular smooth muscle cells. Front Med (Lausanne) 2023; 10:1193660. [PMID: 37469665 PMCID: PMC10352799 DOI: 10.3389/fmed.2023.1193660] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/12/2023] [Indexed: 07/21/2023] Open
Abstract
Atherosclerosis (AS) is characterized by the accumulation of lipids, fibrous elements, and calcification in the innermost layers of arteries. Vascular calcification (VC), the deposition of calcium and phosphate within the arterial wall, is an important characteristic of AS natural history. However, medial arterial calcification (MAC) differs from intimal calcification and cannot simply be explained as the consequence of AS. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are directly involved in AS and VC processes. Understanding the communication between ECs and VSMCs is critical in revealing mechanisms underlying AS and VC. Extracellular vesicles (EVs) are found as intercellular messengers in kinds of physiological processes and pathological progression. Non-coding RNAs (ncRNAs) encapsulated in EVs are involved in AS and VC, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). The effects of ncRNAs have not been comprehensively understood, especially encapsulated in EVs. Some ncRNAs have demonstrated significant roles in AS and VC, but it remains unclear the functions of the majority ncRNAs detected in EVs. In this review, we summarize ncRNAs encapsulated in EC-EVs and VSMC-EVs, and the signaling pathways that are involved in AS and VC.
Collapse
Affiliation(s)
- Fengyi Yu
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yingjie Duan
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chongmei Liu
- Department of Pathology, Yueyang People's Hospital, Yueyang, Hunan, China
| | - Hong Huang
- Hengyang Medical School, The First Affiliated Hospital, Institute of Clinical Medicine, University of South China, Hengyang, Hunan, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhangxiu He
- Department of Nephrology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
8
|
Fernández-Villabrille S, Martín-Carro B, Martín-Vírgala J, Alonso-Montes C, Fernández-Fernández A, Martínez-Salgado C, Fernández-Martín JL, Naves-Díaz M, Cannata-Andía JB, Carrillo-López N, Panizo S. Phosphorus May Induce Phenotypic Transdifferentiation of Vascular Smooth Muscle Cells through the Reduction of microRNA-145. Nutrients 2023; 15:2918. [PMID: 37447244 DOI: 10.3390/nu15132918] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Phosphorus is a vital element for life found in most foods as a natural component, but it is also one of the most used preservatives added during food processing. High serum phosphorus contributes to develop vascular calcification in chronic kidney disease; however, it is not clear its effect in a population without kidney damage. The objective of this in vivo and in vitro study was to investigate the effect of high phosphorus exposure on the aortic and serum levels of miR-145 and its effect on vascular smooth muscle cell (VSMCs) changes towards less contractile phenotypes. The study was performed in aortas and serum from rats fed standard and high-phosphorus diets, and in VSMCs exposed to different concentrations of phosphorus. In addition, miR-145 silencing and overexpression experiments were carried out. In vivo results showed that in rats with normal renal function fed a high P diet, a significant increase in serum phosphorus was observed which was associated to a significant decrease in the aortic α-actin expression which paralleled the decrease in aortic and serum miR-145 levels, with no changes in the osteogenic markers. In vitro results using VSMCs corroborated the in vivo findings. High phosphorus first reduced miR-145, and afterwards α-actin expression. The miR-145 overexpression significantly increased α-actin expression and partially prevented the increase in calcium content. These results suggest that miR-145 could be an early biomarker of vascular calcification, which could give information about the initiation of the transdifferentiation process in VSMCs.
Collapse
Affiliation(s)
- Sara Fernández-Villabrille
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
| | - Beatriz Martín-Carro
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
| | - Julia Martín-Vírgala
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
| | - Cristina Alonso-Montes
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
| | | | - Carlos Martínez-Salgado
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José L Fernández-Martín
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
| | - Manuel Naves-Díaz
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Jorge B Cannata-Andía
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
- Department of Medicine, Universidad de Oviedo, 33011 Oviedo, Spain
| | - Natalia Carrillo-López
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
| | - Sara Panizo
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain
| |
Collapse
|
9
|
Ding N, Lv Y, Su H, Wang Z, Kong X, Zhen J, Lv Z, Wang R. Vascular calcification in CKD: New insights into its mechanisms. J Cell Physiol 2023; 238:1160-1182. [PMID: 37269534 DOI: 10.1002/jcp.31021] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/28/2023] [Indexed: 06/05/2023]
Abstract
Vascular calcification (VC) is a common complication of chronic kidney disease (CKD) and contributes to an increased risk of cardiovascular morbidity and mortality. However, effective therapies are still unavailable at present. It has been well established that VC associated with CKD is not a passive process of calcium phosphate deposition, but an actively regulated and cell-mediated process that shares many similarities with bone formation. Additionally, numerous studies have suggested that CKD patients have specific risk factors and contributors to the development of VC, such as hyperphosphatemia, uremic toxins, oxidative stress and inflammation. Although research efforts in the past decade have greatly improved our knowledge of the multiple factors and mechanisms involved in CKD-related VC, many questions remain unanswered. Moreover, studies from the past decade have demonstrated that epigenetic modifications abnormalities, such as DNA methylation, histone modifications and noncoding RNAs, play an important role in the regulation of VC. This review seeks to provide an overview of the pathophysiological and molecular mechanisms of VC associated with CKD, mainly focusing on the involvement of epigenetic modifications in the initiation and progression of uremic VC, with the aim to develop promising therapies for CKD-related cardiovascular events in the future.
Collapse
Affiliation(s)
- Nannan Ding
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yaodong Lv
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Hong Su
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ziyang Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xianglei Kong
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Junhui Zhen
- Department of Pathology, Shandong University, Jinan, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
10
|
Serum and Urinary Soluble α-Klotho as Markers of Kidney and Vascular Impairment. Nutrients 2023; 15:nu15061470. [PMID: 36986200 PMCID: PMC10057552 DOI: 10.3390/nu15061470] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/22/2023] Open
Abstract
This study was designed to investigate the controversy on the potential role of sKlotho as an early biomarker in Chronic Kidney Disease–Mineral Bone Disorder (CKD-MBD), to assess whether sKlotho is a reliable marker of kidney α-Klotho, to deepen the effects of sKlotho on vascular smooth muscle cells (VSMCs) osteogenic differentiation and to evaluate the role of autophagy in this process. Experimental studies were conducted in CKD mice fed a normal phosphorus (CKD+NP) or high phosphorus (CKD+HP) diet for 14 weeks. The patients’ study was performed in CKD stages 2–5 and in vitro studies which used VSMCs exposed to non-calcifying medium or calcifying medium with or without sKlotho. The CKD experimental model showed that the CKD+HP group reached the highest serum PTH, P and FGF23 levels, but the lowest serum and urinary sKlotho levels. In addition, a positive correlation between serum sKlotho and kidney α-Klotho was found. CKD mice showed aortic osteogenic differentiation, together with increased autophagy. The human CKD study showed that the decline in serum sKlotho is previous to the rise in FGF23. In addition, both serum sKlotho and FGF23 levels correlated with kidney function. Finally, in VSMCs, the addition of sKlotho prevented osteogenic differentiation and induced autophagy. It can be concluded that serum sKlotho was the earliest CKD-MBD biomarker, a reliable indicator of kidney α-Klotho and that might protect against osteogenic differentiation by increasing autophagy. Nevertheless, further studies are needed to investigate the mechanisms of this possible protective effect.
Collapse
|
11
|
Zhao H, Liu H, Liu Y, Jin J, He Q, Lin B. The role of extracellular vesicles in vascular calcification in chronic kidney disease. Front Med (Lausanne) 2022; 9:997554. [PMID: 36388921 PMCID: PMC9651939 DOI: 10.3389/fmed.2022.997554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/10/2022] [Indexed: 09/08/2024] Open
Abstract
Widespread vascular calcification (VC) in patients with chronic kidney disease (CKD) is the pathological basis for the development of cardiovascular disease, and VC has been identified as an independent risk factor for increased cardiovascular mortality in cases of CKD. While VC was earlier thought to be a passive deposition process following calcium and phosphorus supersaturation, recent studies have suggested that it is an active, modifiable, biological process similar to bone development. The involvement of extracellular vesicles (EVs) in the process of VC has been reported as an important transporter of material transport and intercellular communication. This paper reviews the mechanism of the role of EVs, especially exosomes, in VC and the regulation of VC by stem cell-derived EVs, and discusses the possible and promising application of related therapeutic targets in the clinical setting.
Collapse
Affiliation(s)
- Huan Zhao
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| | - Haojie Liu
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| | - Yueming Liu
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| | - Juan Jin
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| | - Qiang He
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bo Lin
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejian, China
| |
Collapse
|
12
|
Ye YC, Chang ZH, Wang P, Wang YW, Liang J, Chen C, Wang JJ, Sun HT, Wang Y, Li XH. Infarct-preconditioning exosomes of umbilical cord mesenchymal stem cells promoted vascular remodeling and neurological recovery after stroke in rats. Stem Cell Res Ther 2022; 13:378. [PMID: 35902882 PMCID: PMC9330700 DOI: 10.1186/s13287-022-03083-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/21/2022] [Indexed: 12/30/2022] Open
Abstract
Background Stroke is the leading cause of disability worldwide, resulting in severe damage to the central nervous system and disrupting neurological functions. There is no effective therapy for promoting neurological recovery. Growing evidence suggests that the composition of exosomes from different microenvironments may benefit stroke. Therefore, it is reasonable to assume that exosomes secreted in response to infarction microenvironment could have further therapeutic effects. Methods In our study, cerebral infarct tissue extracts were used to pretreat umbilical cord mesenchymal stem cells (UCMSC). Infarct-preconditioned exosomes were injected into rats via tail vein after middle cerebral artery occlusion (MCAO). The effect of infarct-preconditioned exosomes on the neurological recovery of rats was examined using Tunel assay, 2,3,5-triphenyltetrazolium chloride (TTC) assay, magnetic resonance imaging (MRI) analyses, modified Neurological Severity Score (mNSS), Morris water maze (MWM), and vascular remodeling analysis. Mi-RNA sequencing and functional enrichment analysis were used to validate the signal pathway involved in the effect of infarct-preconditioned exosomes. Human umbilical vein endothelial cells (HUVECs) were co-cultured with the isolated exosomes. Cell Counting Kit-8 (CCK-8) assay, scratch healing, and Western blot analysis were used to detect the biological behavior of HUVECs. Results The results showed that compared with normal exosomes, infarct-preconditioned exosomes further promoted vascular remodeling and recovery of neurological function after stroke. The function of upregulated miRNAs and their target genes which is beneficial to vascular smooth muscle cells verified the importance of vascular remodeling in improving stroke. Better resistance to oxygen–glucose deprivation/reoxygenation (OGD/R), reduced apoptosis, and enhanced migration were observed in infarct-preconditioned exosomes-treated umbilical vein endothelial cells. Conclusions Our results demonstrated that infarct-preconditioned exosomes promoted neurological recovery after stroke by enhancing vascular endothelial remodeling, suggested that infarct-preconditioned exosomes could be a novel way to alleviate brain damage following a stroke. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03083-9.
Collapse
Affiliation(s)
- Yi-Chao Ye
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Peng Wang
- Health Management Department, Tianjin Hospital, Tianjin University, Tianjin, 300299, China
| | - You-Wei Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jun Liang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Yi Wang
- Neurology Department, Tianjin Hospital, Tianjin University, Tianjin, 300299, China.
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
13
|
Wu YY, Shan SK, Lin X, Xu F, Zhong JY, Wu F, Duan JY, Guo B, Li FXZ, Wang Y, Zheng MH, Xu QS, Lei LM, Ou-Yang WL, Tang KX, Li CC, Ullah MHE, Yuan LQ. Cellular Crosstalk in the Vascular Wall Microenvironment: The Role of Exosomes in Vascular Calcification. Front Cardiovasc Med 2022; 9:912358. [PMID: 35677687 PMCID: PMC9168031 DOI: 10.3389/fcvm.2022.912358] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/02/2022] [Indexed: 07/20/2023] Open
Abstract
Vascular calcification is prevalent in aging, diabetes, chronic kidney disease, cardiovascular disease, and certain genetic disorders. However, the pathogenesis of vascular calcification is not well-understood. It has been progressively recognized that vascular calcification depends on the bidirectional interactions between vascular cells and their microenvironment. Exosomes are an essential bridge to mediate crosstalk between cells and organisms, and thus they have attracted increased research attention in recent years. Accumulating evidence has indicated that exosomes play an important role in cardiovascular disease, especially in vascular calcification. In this review, we introduce vascular biology and focus on the crosstalk between the different vessel layers and how their interplay controls the process of vascular calcification.
Collapse
Affiliation(s)
- Yun-Yun Wu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yu Zhong
- Department of Nuclear Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Feng Wu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yue Duan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fu-Xing-Zi Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Wang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Lu Ou-Yang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Xin Tang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chang-Chun Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Hasnain Ehsan Ullah
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Peng Z, Duan Y, Zhong S, Chen J, Li J, He Z. RNA-seq analysis of extracellular vesicles from hyperphosphatemia-stimulated endothelial cells provides insight into the mechanism underlying vascular calcification. BMC Nephrol 2022; 23:192. [PMID: 35597927 PMCID: PMC9123672 DOI: 10.1186/s12882-022-02823-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/16/2022] [Indexed: 11/19/2022] Open
Abstract
Background Hyperphosphatemia (HP) is associated with vascular calcification (VC) in chronic kidney disease (CKD). However, relationship between HP-induced-endothelial extracellular vesicles (HP-EC-EVs) and VC is unclear, and miR expression in HP-EC-EVs has not been determined. Methods We isolated HP-EC-EVs from endothelial cells with HP and observed that HP-EC-EVs were up-taken by vascular smooth muscle cells (VSMCs). HP-EC-EVs inducing calcium deposition was characterized by Alizarin Red S, colourimetric analysis and ALP activity. To investigate the mechanism of HP-EC-EVs-induced VSMC calcification, RNA-sequencing for HP-EC-EVs was performed. Results We first demonstrated that HP-EC-EVs induced VSMC calcification in vitro. RNA-seq analysis of HP-EC-EVs illustrated that one known miR (hsa-miR-3182) was statistically up-regulated and twelve miRs were significantly down-regulated, which was verified by qRT-PCR. We predicted 58,209 and 74,469 target genes for those down- and up-regulated miRs respectively through miRDB, miRWalk and miRanda databases. GO terms showed that down- and up-regulated targets were mostly enriched in calcium-dependent cell–cell adhesion via plama membrane cell-adhesion molecules (GO:0,016,338, BP) and cell adhesion (GO:0,007,155, BP), plasma membrane (GO:0,005,886, CC), and metal ion binding (GO:0,046,914, MF) and ATP binding (GO:0,005,524, MF) respectively. Top-20 pathways by KEGG analysis included calcium signaling pathway, cAMP signaling pathway, and ABC transporters, which were closely related to VC. Conclusion Our results indicated that those significantly altered miRs, which were packaged in HP-EC-EVs, may play an important role in VC by regulating related pathways. It may provide novel insight into the mechanism of CKD calcification. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02823-6.
Collapse
Affiliation(s)
- Zhong Peng
- The First Affiliated Hospital, Department of Gastroenterology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yingjie Duan
- The First Affiliated Hospital, Department of Nephrology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shuzhu Zhong
- The First Affiliated Hospital, Department of Nephrology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230002, China
| | - Jianlong Li
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Pediatrics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Zhangxiu He
- The First Affiliated Hospital, Department of Nephrology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
15
|
Liu Q, Qi H, Yao L. A long non-coding RNA H19/microRNA-138/TLR3 network is involved in high phosphorus-mediated vascular calcification and chronic kidney disease. Cell Cycle 2022; 21:1667-1683. [PMID: 35435133 PMCID: PMC9302514 DOI: 10.1080/15384101.2022.2064957] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Vascular calcification, characterized by the accumulation of calcium-phosphate crystals in blood vessels, is a major cause of cardiovascular complications and chronic kidney disease (CKD)-related death. This work focuses on the molecules involved in high-phosphorus-mediated vascular calcification in CKD. A rat model of CKD was established by 5/6 nephrectomy, and the rats were given normal phosphorus diet (NPD) or high phosphorus diet (HPD). HPD decreased kidney function, increased the concentration of calcium ion and damaged vascular structure in the thoracic aorta of diseased rats. A high phosphorus condition enhanced calcium deposition in vascular smooth muscle cells (VSMCs). High phosphorus also increased the expression of RUNX2 whereas reduced the expression of α-SM actin in the aortic tissues and VSMCs. Long non-coding RNA (lncRNA) H19 was upregulated in the aortic tissues after HPD treatment. H19 bound to microRNA (miR)-138 to block its inhibitory effect on TLR3 mRNA and activated the NF-κB signaling pathway. Downregulation of H19 or TLR3 alleviated, whereas downregulation of miR-138 aggravated the calcification and vascular damage in model rats and VSMCs. In conclusion, this study demonstrates that the H19/miR-138/TLR3 axis is involved in high phosphorus-mediated vascular calcification in rats with CKD.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Nephrology, Fuyang Hospital of Anhui Medical University, Fuyang Anhui, P.R. China
| | - Huimeng Qi
- Department of General Practice, Fuyang Hospital of Anhui Medical University, Fuyang Anhui, P.R. China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang Liaoning, P.R. China
| |
Collapse
|
16
|
Lv W, Cui C, Wang Z, Jiang J, Deng B. A High Serum Phosphate and Calcium-Phosphate Product Is Associated With Cerebral Small Vascular Disease in Patients With Stroke: A Real-World Study. Front Nutr 2022; 9:801667. [PMID: 35445062 PMCID: PMC9013770 DOI: 10.3389/fnut.2022.801667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 03/04/2022] [Indexed: 01/23/2023] Open
Abstract
Cerebral small vessel disease (CSVD) is a slowly progressive disease, often accompanied by stroke, and results in dementia, depression, and cognitive impairment. It was already known that calcium and phosphorus metabolism (CPM) disorders were associated with vascular-related adverse events. The risk factors of CSVD and the relationship between serum calcium (Ca), phosphorus (P), calcium-phosphate product (Ca × P), and CSVD in patients with stroke without CPM disorders are still obscure. In our study, 528 patients with stroke without CPM disorders were enrolled in a cohort from a consecutive hospital-based stroke registry, with 488 patients with CSVD as cases and 140 without CSVD as controls. The patients with CSVD were further sub-grouped into lacunes, white matter hyperintensities (WMHs), and cerebral microbleeds (CMBs). By applying univariate and multivariate logistic regression analysis, the following novel findings were obtained: (i) up to 76.19% of patients with stroke had signs of CSVD, and lacunes are the most common subtype. Notably, 22.96% of patients with CSVD had multiple subtypes coexisted. (ii) Compared with patients without CSVD, patients with CSVD had higher levels of age, rate of hypertension or diabetes, serum Ca, P, Ca × P, and lower levels of white blood cell (WBC) and hemoglobin (HB). (iii) We developed 2 predictive models and nomograms for predicting CSVD, in addition to the known factors (age and hypertension). The levels of P and Ca × P were positively correlated with the risk of CSVD (P: OR = 3,720.401, 95% CI (646.665–21,404.249); Ca × P: OR = 1.294, 95% CI (1.222–1.370)). (iv) The models were further validated in subtypes of CSVD, including lacunes, WMHs, and CMBs, and the results were still valid among the subtypes. In summary, CSVD was highly prevalent in patients with stroke, and high serum P and Ca × P are potential risk factors of CSVD and all subtypes including lacunes, WMHs, and CMBs.
Collapse
Affiliation(s)
- Wenjing Lv
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Can Cui
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zixuan Wang
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Junqi Jiang
- Medical College, Qingdao University, Qingdao, China
| | - Binbin Deng
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Binbin Deng
| |
Collapse
|
17
|
MicroRNAs in Valvular Heart Diseases: Biological Regulators, Prognostic Markers and Therapeutical Targets. Int J Mol Sci 2021; 22:ijms222212132. [PMID: 34830016 PMCID: PMC8618095 DOI: 10.3390/ijms222212132] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022] Open
Abstract
miRNAs have recently attracted investigators’ interest as regulators of valvular diseases pathogenesis, diagnostic biomarkers, and therapeutical targets. Evidence from in-vivo and in-vitro studies demonstrated stimulatory or inhibitory roles in mitral valve prolapse development, aortic leaflet fusion, and calcification pathways, specifically osteoblastic differentiation and transcription factors modulation. Tissue expression assessment and comparison between physiological and pathological phenotypes of different disease entities, including mitral valve prolapse and mitral chordae tendineae rupture, emerged as the best strategies to address miRNAs over or under-representation and thus, their impact on pathogeneses. In this review, we discuss the fundamental intra- and intercellular signals regulated by miRNAs leading to defects in mitral and aortic valves, congenital heart diseases, and the possible therapeutic strategies targeting them. These miRNAs inhibitors are comprised of antisense oligonucleotides and sponge vectors. The miRNA mimics, miRNA expression vectors, and small molecules are instead possible practical strategies to increase specific miRNA activity. Advantages and technical limitations of these new drugs, including instability and complex pharmacokinetics, are also presented. Novel delivery strategies, such as nanoparticles and liposomes, are described to improve knowledge on future personalized treatment directions.
Collapse
|
18
|
Cannata-Andía JB, Carrillo-López N, Messina OD, Hamdy NAT, Panizo S, Ferrari SL. Pathophysiology of Vascular Calcification and Bone Loss: Linked Disorders of Ageing? Nutrients 2021; 13:3835. [PMID: 34836090 PMCID: PMC8623966 DOI: 10.3390/nu13113835] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
Vascular Calcification (VC), low bone mass and fragility fractures are frequently observed in ageing subjects. Although this clinical observation could be the mere coincidence of frequent age-dependent disorders, clinical and experimental data suggest that VC and bone loss could share pathophysiological mechanisms. Indeed, VC is an active process of calcium and phosphate precipitation that involves the transition of the vascular smooth muscle cells (VSMCs) into osteoblast-like cells. Among the molecules involved in this process, parathyroid hormone (PTH) plays a key role acting through several mechanisms which includes the regulation of the RANK/RANKL/OPG system and the Wnt/ß-catenin pathway, the main pathways for bone resorption and bone formation, respectively. Furthermore, some microRNAs have been implicated as common regulators of bone metabolism, VC, left ventricle hypertrophy and myocardial fibrosis. Elucidating the common mechanisms between ageing; VC and bone loss could help to better understand the potential effects of osteoporosis drugs on the CV system.
Collapse
Affiliation(s)
- Jorge B. Cannata-Andía
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (N.C.-L.); (S.P.)
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (N.C.-L.); (S.P.)
| | - Osvaldo D. Messina
- Investigaciones Reumatológicas y Osteológicas (IRO), Buenos Aires 1114, Argentina;
| | - Neveen A. T. Hamdy
- Center for Bone Quality, Division Endocrinology, Department of Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Sara Panizo
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (N.C.-L.); (S.P.)
| | - Serge L. Ferrari
- Service and Laboratory of Bone Diseases, Department of Medicine, Faculty of Medicine, Geneva University Hospital, 1211 Geneva, Switzerland;
| | | |
Collapse
|
19
|
Ferreira TJ, de Araújo CC, Lima ACDS, Matida LM, Griebeler AFM, Coelho ASG, Gontijo APM, Cominetti C, Vêncio EF, Horst MA. Dietary Intake is Associated with miR-31 and miR-375 Expression in Patients with Head and Neck Squamous Cell Carcinoma. Nutr Cancer 2021; 74:2049-2058. [PMID: 34647497 DOI: 10.1080/01635581.2021.1990972] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
MicroRNAs (miRNAs) are important epigenetic regulators in head and neck squamous cell carcinoma (HNSCC), with miR-31 being considered an oncomir and miR-375, a tumor suppressor miR, which are up- and down-regulated in HNSCC, respectively. Nutrients are known to influence miRNA expression; however, this association is poorly explored in HNSCC. This work aimed to identify associations between dietary intake and the expression of miR-31 and miR-375 in patients newly diagnosed with HNSCC. The expression of miR-31 was positively associated with the consumption of iron (β = 16.65) and vitamin C (β = 0.37), and inversely associated with total sugar (β = -0.88), cholesterol (β= -0.23), vitamin B9 (β= -0.37) and zinc (β = -5.66) intake. The expression of miR-375 was positively associated with the consumption of selenium (β = 1.52), vitamin C (β = 0.17) and vitamin D (β = 13.01), and inversely associated with the consumption of added sugar (β = -0.49), phosphorus (β= -0.27) and vitamin B12 (β = -10.80). Our findings showed important associations between dietary intake and miR-31 and miR-375 expression in HNSCC, offering possible directions for further studies investigating how nutrients interfere with carcinogenesis.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.1990972 .
Collapse
Affiliation(s)
- Tathiany Jéssica Ferreira
- Nutritional Genomics Research Group, School of Nutrition, Federal University of Goias, Goiania, Brazil
| | - Caroline Castro de Araújo
- Nutritional Genomics Research Group, School of Nutrition, Federal University of Goias, Goiania, Brazil
| | | | - Larissa Morinaga Matida
- Nutritional Genomics Research Group, School of Nutrition, Federal University of Goias, Goiania, Brazil
| | | | | | | | - Cristiane Cominetti
- Nutritional Genomics Research Group, School of Nutrition, Federal University of Goias, Goiania, Brazil
| | | | - Maria Aderuza Horst
- Nutritional Genomics Research Group, School of Nutrition, Federal University of Goias, Goiania, Brazil
| |
Collapse
|
20
|
Nguyen DDN, Zain SM, Kamarulzaman MH, Low TY, Chilian WM, Pan Y, Ting KN, Hamid A, Abdul Kadir A, Pung YF. Intracellular and exosomal microRNAome profiling of human vascular smooth muscle cells during replicative senescence. Am J Physiol Heart Circ Physiol 2021; 321:H770-H783. [PMID: 34506226 DOI: 10.1152/ajpheart.00058.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular aging is highly associated with cardiovascular morbidity and mortality. Although the senescence of vascular smooth muscle cells (VSMCs) has been well established as a major contributor to vascular aging, intracellular and exosomal microRNA (miRNA) signaling pathways in senescent VSMCs have not been fully elucidated. This study aimed to identify the differential expression of intracellular and exosomal miRNA in human VSMCs (hVSMCs) during replicative senescence. To achieve this aim, intracellular and exosomal miRNAs were isolated from hVSMCs and subsequently subjected to whole genome small RNA next-generation sequencing, bioinformatics analyses, and qPCR validation. Three significant findings were obtained. First, senescent hVSMC-derived exosomes tended to cluster together during replicative senescence and the molecular weight of the exosomal protein tumor susceptibility gene 101 (TSG-101) increased relative to the intracellular TSG-101, suggesting potential posttranslational modifications of exosomal TSG-101. Second, there was a significant decrease in both intracellular and exosomal hsa-miR-155-5p expression [n = 3, false discovery rate (FDR) < 0.05], potentially being a cell type-specific biomarker of hVSMCs during replicative senescence. Importantly, hsa-miR-155-5p was found to associate with cell-cycle arrest and elevated oxidative stress. Lastly, miRNAs from the intracellular pool, that is, hsa-miR-664a-3p, hsa-miR-664a-5p, hsa-miR-664b-3p, hsa-miR-4485-3p, hsa-miR-10527-5p, and hsa-miR-12136, and that from the exosomal pool, that is, hsa-miR-7704, were upregulated in hVSMCs during replicative senescence (n = 3, FDR < 0.05). Interestingly, these novel upregulated miRNAs were not functionally well annotated in hVSMCs to date. In conclusion, hVSMC-specific miRNA expression profiles during replicative senescence potentially provide valuable insights into the signaling pathways leading to vascular aging.NEW & NOTEWORTHY This is the first study on intracellular and exosomal miRNA profiling on human vascular smooth muscle cells during replicative senescence. Specific dysregulated sets of miRNAs were identified from human vascular smooth muscle cells. Hsa-miR-155-5p was significantly downregulated in both intracellular and exosomal hVSMCs, suggesting its crucial role in cellular senescence. Hsa-miR-155-5p might be the mediator in linking cellular senescence to vascular aging and atherosclerosis.
Collapse
Affiliation(s)
- Diem Duong Ngoc Nguyen
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Shamsul Mohd Zain
- The Pharmacogenomics Laboratory, Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Teck Yew Low
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - William M Chilian
- Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Yan Pan
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Kang Nee Ting
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Aini Hamid
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Arifah Abdul Kadir
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, University Putra Malaysia, Selangor, Malaysia
| | - Yuh-Fen Pung
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| |
Collapse
|
21
|
He L, Xu J, Bai Y, Zhang H, Zhou W, Cheng M, Zhang D, Zhang L, Zhang S. MicroRNA-103a regulates the calcification of vascular smooth muscle cells by targeting runt-related transcription factor 2 in high phosphorus conditions. Exp Ther Med 2021; 22:1036. [PMID: 34373722 PMCID: PMC8343701 DOI: 10.3892/etm.2021.10468] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
Vascular calcification, such as atherosclerosis, is a serious complication of chronic kidney disease that is characterized by tunica media calcification, and has gained increasing attention from researchers. The commonly observed association between vascular calcification and osteoporosis suggests a link between bone and vascular disorders. As microRNAs (miRNAs) have a wide range of gene regulation functions, such as cell proliferation, apoptosis, stress and transdifferentiation, the current study aimed to determine whether miRNAs play a vital role in the calcification and osteoblastic differentiation of rat thoracic aorta vascular smooth muscle cells (VSMCs). Gene expression analysis was performed on seven miRNAs (miR-29a, -30b, -103a, -125b, -133a, -143 and -211) that maybe potentially involved in the differentiation of smooth muscle cells into osteoblastic cells. The results showed that the levels of miR-29a, -30b, -103a, -125b and -143 were markedly reduced in the VSMC calcification model, particularly miR-103a, whereas runt-related transcription factor 2 (RUNX2) expression was increased. Furthermore, it was found that the expression of RUNX2 was significantly decreased following the upregulation of miR-103a, and that the expression of RUNX2 was significantly increased by downregulating miR-103a in VSMCs. Therefore, it was concluded that miR-103a plays a notable role in the transdifferentiation of the VSMCs in high phosphorus-induced calcification by targeting the regulation of RUNX2, and may therefore constitute a new target for the diagnosis and treatment of vascular calcification.
Collapse
Affiliation(s)
- Lei He
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| | - Jinsheng Xu
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| | - Yaling Bai
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| | - Huiran Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| | - Wei Zhou
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| | - Meijuan Cheng
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| | - Dongxue Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| | - Lu Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| | - Shenglei Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
22
|
Zhang Z, Jiang W, Hu M, Gao R, Zhou X. MiR-486-3p promotes osteogenic differentiation of BMSC by targeting CTNNBIP1 and activating the Wnt/β-catenin pathway. Biochem Biophys Res Commun 2021; 566:59-66. [PMID: 34118593 DOI: 10.1016/j.bbrc.2021.05.098] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/29/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Dysfunction in the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) leads to bone loss/osteoporosis. The catenin beta interacting protein 1 (CTNNBIP1) is an inhibitor of Wnt/β-catenin signaling, whose role in osteogenesis remains elusive. This study aimed to reveal the effects of miR-486-3p/CTNNBIP1 in osteogenesis. METHODS Bone marrow samples from healthy individuals and osteoporosis patients and mice with sham or ovariectomy (OVX) surgeries were collected. Levels of CTNNBIP1 and miR-486-3p were assessed. A dual-luciferase reporter assay was used to confirm the interactions between CTNNBIP1 and miR-486-3p. MiR-486-3p mimics/inhibitor or CTNNBIP1 overexpression lentiviruses were transfected to human BMSCs (hBMSCs) and an osteogenic assay was performed. Alizarin red S (ARS) and Alkaline phosphatase (ALP) intensity and expression of osteogenic genes Runx2, Alp, Cola1 and Bglap were measured. Key proteins in the Wnt/β-catenin pathway including active β-catenin, Bcl-2, and Cyclin D1 were assessed. RESULTS CTNNBIP1 was upregulated while miR-486-3p was downregulated in osteoporosis patients and OVX mice. CTNNBIP1 was confirmed as a target of miR-486-3p. MiR-486-3p overexpression promoted, while miR-486-3p knockdown suppressed, osteogenic differentiation and Wnt/β-catenin signaling. Rescue experiments confirmed the negative effects of CTNNBIP1 overexpression on osteoblastic differentiation and that miR-486-3p mimics could reverse canonical Wnt signaling. CONCLUSION This study demonstrated that miR-486-3p targets CTNNBIP1, thus activating the Wnt/β-catenin signaling pathway to promote osteogenesis of BMSCs.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, PR China; College of Basic Medicine, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, PR China
| | - Weiwei Jiang
- Department of Critical Care Medicine, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, PR China
| | - Miao Hu
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, PR China; College of Basic Medicine, Second Military Medical University, 800 Xiangyin Road, Shanghai, 200433, PR China
| | - Rui Gao
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, PR China
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, PR China.
| |
Collapse
|
23
|
Zhang Y, Wang H, Xia Y. The expression of miR-211-5p in atherosclerosis and its influence on diagnosis and prognosis. BMC Cardiovasc Disord 2021; 21:371. [PMID: 34340677 PMCID: PMC8330028 DOI: 10.1186/s12872-021-02187-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The purpose of this study was to evaluate the diagnostic and prognostic significance of miR-211-5p in atherosclerosis (AS) by detecting the expression level in serum of patients with AS. METHODS A total of 85 healthy controls and 90 asymptomatic AS patients participated in this study. The expression level of miR-211-5p in all subjects were measured by qRT-PCR. Spearman correlation coefficient was used to evaluate the correlation of miR-211-5p with CRP and CIMT. The ROC curve was established to assess the diagnostic value of miR-211-5p in AS. The Kaplan-Meier survival curve and multivariate COX regression analysis were used to evaluate the prognostic significance of miR-211-5p in AS. RESULTS The expression levels of miR-211-5p in AS patients were significantly lower than in healthy controls (P < 0.001), and miR-211-5p showed a significant negative correlation with CRP (r = - 0.639, P < 0.001) and CIMT (r = - 0.730, P < 0.001). The AUC of the ROC curve was 0.900, the specificity and the sensitivity were 84.7% and 78.9%, respectively, which indicating that miR-211-5p had diagnostic value for AS. Survival analysis showed that patients with low miR-211-5p expression were more likely to have cardiovascular end-point events (Log rank P = 0.013). CONCLUSION Serum miR-211-5p could be used as a new biomarker for the diagnosis of AS, and the low expression of miR-211-5p is associated with the poor prognosis of AS.
Collapse
Affiliation(s)
- Yanxia Zhang
- Department of Health Comprehensive Geriatrics, Yidu Central Hospital of Weifang, No. 4138, Linglongshan Road, Weifang, 262500, Shandong, China.
| | - Huiyun Wang
- Department of Health Comprehensive Geriatrics, Yidu Central Hospital of Weifang, No. 4138, Linglongshan Road, Weifang, 262500, Shandong, China
| | - Yu Xia
- Department of Health Comprehensive Geriatrics, Yidu Central Hospital of Weifang, No. 4138, Linglongshan Road, Weifang, 262500, Shandong, China
| |
Collapse
|
24
|
Chao CT, Yeh HY, Tsai YT, Chiang CK, Chen HW. A combined microRNA and target protein-based panel for predicting the probability and severity of uraemic vascular calcification: a translational study. Cardiovasc Res 2021; 117:1958-1973. [PMID: 32866261 DOI: 10.1093/cvr/cvaa255] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/24/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Vascular calcification (VC) increases the future risk of cardiovascular events in uraemic patients, but effective therapies are still unavailable. Accurate identification of those at risk of developing VC using pathogenesis-based biomarkers is of particular interest and may facilitate individualized risk stratification. We aimed to uncover microRNA (miRNA)-target protein-based biomarker panels for evaluating uraemic VC probability and severity. METHODS AND RESULTS We created a three-tiered in vitro VC model and an in vivo uraemic rat model receiving high phosphate diet to mimic uraemic VC. RNAs from the three-tiered in vitro and in vivo uraemic VC models underwent miRNA and mRNA microarray, with results screened for differentially expressed miRNAs and their target genes as biomarkers. Findings were validated in original models and additionally in an ex vivo VC model and human cells, followed by functional assays of identified miRNAs and target proteins, and tests of sera from end-stage renal disease (ESRD) and non-dialysis-dependent chronic kidney disease (CKD) patients without and with VC. Totally 122 down-regulated and 119 up-regulated miRNAs during calcification progression were identified initially; further list narrowing based on miRNA-mRNA pairing, anti-correlation, and functional enrichment left 16 and 14 differentially expressed miRNAs and mRNAs. Levels of four miRNAs (miR-10b-5p, miR-195, miR-125b-2-3p, and miR-378a-3p) were shown to decrease throughout all models tested, while one mRNA (SULF1, a potential target of miR-378a-3p) exhibited the opposite trend concurrently. Among 96 ESRD (70.8% with VC) and 59 CKD patients (61% with VC), serum miR-125b2-3p and miR-378a-3p decreased with greater VC severity, while serum SULF1 levels increased. Adding serum miR-125b-2-3p, miR-378a-3p, and SULF1 into regression models for VC substantially improved performance compared to using clinical variables alone. CONCLUSION Using a translational approach, we discovered a novel panel of biomarkers for gauging the probability/severity of uraemic VC based on miRNAs/target proteins, which improved the diagnostic accuracy.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Biomarkers/blood
- Cells, Cultured
- Disease Models, Animal
- Female
- Gene Expression Profiling
- Gene Regulatory Networks
- Humans
- Male
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Organ Culture Techniques
- Predictive Value of Tests
- Protein Interaction Maps
- Proteome
- Proteomics
- Rats, Sprague-Dawley
- Risk Assessment
- Risk Factors
- Severity of Illness Index
- Signal Transduction
- Sulfotransferases/blood
- Transcriptome
- Translational Research, Biomedical
- Uremia/complications
- Uremia/genetics
- Uremia/metabolism
- Vascular Calcification/etiology
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Rats
Collapse
Affiliation(s)
- Chia-Ter Chao
- Division of Nephrology, Department of Medicine, National Taiwan University Hospital Bei-Hu Branch, No. 87, Neijiang Street, Wanhua District, Taipei 10845, Taiwan
- Graduate Institute of Toxicology, National Taiwan University, College of Medicine, No.1, Section 4, Ren-Ai Road, Zhongzheng District, Taipei 10051, Taiwan
- Department of Internal Medicine, National Taiwan University College of Medicine, No.1, Section 4, Ren-Ai Road, Zhongzheng District, Taipei 10051, Taiwan
| | - Hsiang-Yuan Yeh
- School of Big Data Management, Soochow University, No.70, Linxi Road, Shilin District, Taipei 11102, Taiwan
| | - You-Tien Tsai
- Division of Nephrology, Department of Medicine, National Taiwan University Hospital Bei-Hu Branch, No. 87, Neijiang Street, Wanhua District, Taipei 10845, Taiwan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, National Taiwan University, College of Medicine, No.1, Section 4, Ren-Ai Road, Zhongzheng District, Taipei 10051, Taiwan
- Department of Integrative Diagnostics and Therapeutics, National Taiwan University Hospital, No. 7, Zhongshan South Road, Zhongzheng District, Taipei 10002, Taiwan
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, National Taiwan University, College of Medicine, No.1, Section 4, Ren-Ai Road, Zhongzheng District, Taipei 10051, Taiwan
| |
Collapse
|
25
|
Song N, Luo J, Huang L, Tian H, Chen Y, He Q. miR-204-5p and miR-211 Synergistically Downregulate the α S1-Casein Content and Contribute to the Lower Allergy of Goat Milk. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:5353-5362. [PMID: 33939400 DOI: 10.1021/acs.jafc.1c01147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
αS1-Casein (encoded by the CSN1S1 gene) is associated with higher rates of allergy than other milk protein components for humans. microRNAs (miRNAs) as small noncoding RNA molecules regulate gene expression and influence diverse biological processes. However, little is known about the regulation of milk protein synthesis by miRNAs in ruminants. In this study, we aim to investigate the regulatory roles of miR-204 family members (miR-204-5p and miR-211) on αS1-casein in goat mammary epithelial cells (GMEC). Here, we observed that the CSN1S1 mRNA level is upregulated, while miR-204-5p and miR-211 (miR-204-5p/-211) abundance is downregulated during peak lactation compared with middle lactation of dairy goats. We found that miR-204-5p/-211 synergistically inhibit αS1-casein expression via directly binding to the 3'-untranslated region (3'UTR) of CSN1S1 in GMEC. miR-204-5p/-211 increase β-casein mRNA (CSN2) and protein abundance, as well as the signal transducer and activator of transcription 5a (STAT5a) activity. Further, miR-204-5p/-211 enhance β-casein expression via the CSN1S1-STAT5a signaling axis and promote β-casein transcription by activating the STAT5 response element located in the CSN2 promoter. In conclusion, miR-204-5p/-211 regulate αS1-casein and β-casein synthesis via targeting CSN1S1 in GMEC, which provide the strategy for manipulating miR-204 family members to reduce milk allergy potential and improve ruminant milk quality for human consumption.
Collapse
Affiliation(s)
- Ning Song
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, P. R. China
| | - Jun Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, P. R. China
| | - Lian Huang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, P. R. China
| | - Huibin Tian
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, P. R. China
| | - Yating Chen
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, P. R. China
| | - Qiuya He
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, P. R. China
| |
Collapse
|
26
|
Carrillo-López N, Martínez-Arias L, Alonso-Montes C, Martín-Carro B, Martín-Vírgala J, Ruiz-Ortega M, Fernández-Martín JL, Dusso AS, Rodriguez-García M, Naves-Díaz M, Cannata-Andía JB, Panizo S. The receptor activator of nuclear factor κΒ ligand receptor leucine-rich repeat-containing G-protein-coupled receptor 4 contributes to parathyroid hormone-induced vascular calcification. Nephrol Dial Transplant 2021; 36:618-631. [PMID: 33367746 DOI: 10.1093/ndt/gfaa290] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In chronic kidney disease, serum phosphorus (P) elevations stimulate parathyroid hormone (PTH) production, causing severe alterations in the bone-vasculature axis. PTH is the main regulator of the receptor activator of nuclear factor κB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, which is essential for bone maintenance and also plays an important role in vascular smooth muscle cell (VSMC) calcification. The discovery of a new RANKL receptor, leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), which is important for osteoblast differentiation but with an unknown role in vascular calcification (VC), led us to examine the contribution of LGR4 in high P/high PTH-driven VC. METHODS In vivo studies were conducted in subtotally nephrectomized rats fed a normal or high P diet, with and without parathyroidectomy (PTX). PTX rats were supplemented with PTH(1-34) to achieve physiological serum PTH levels. In vitro studies were performed in rat aortic VSMCs cultured in control medium, calcifying medium (CM) or CM plus 10-7 versus 10-9 M PTH. RESULTS Rats fed a high P diet had a significantly increased aortic calcium (Ca) content. Similarly, Ca deposition was higher in VSMCs exposed to CM. Both conditions were associated with increased RANKL and LGR4 and decreased OPG aorta expression and were exacerbated by high PTH. Silencing of LGR4 or parathyroid hormone receptor 1 (PTH1R) attenuated the high PTH-driven increases in Ca deposition. Furthermore, PTH1R silencing and pharmacological inhibition of protein kinase A (PKA), but not protein kinase C, prevented the increases in RANKL and LGR4 and decreased OPG. Treatment with PKA agonist corroborated that LGR4 regulation is a PTH/PKA-driven process. CONCLUSIONS High PTH increases LGR4 and RANKL and decreases OPG expression in the aorta, thereby favouring VC. The hormone's direct pro-calcifying actions involve PTH1R binding and PKA activation.
Collapse
Affiliation(s)
- Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Cristina Alonso-Montes
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Beatriz Martín-Carro
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Julia Martín-Vírgala
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Marta Ruiz-Ortega
- Vascular and Renal Laboratory Fundación Jimenez Díaz, Universidad Autónoma Madrid, REDinREN-ISCIII, Madrid, Spain
| | - José Luis Fernández-Martín
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Adriana S Dusso
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Minerva Rodriguez-García
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain.,Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain.,Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Sara Panizo
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| |
Collapse
|
27
|
Wang J, Gao Z, Gao P. MiR-133b Modulates the Osteoblast Differentiation to Prevent Osteoporosis Via Targeting GNB4. Biochem Genet 2021; 59:1146-1157. [PMID: 33687637 DOI: 10.1007/s10528-021-10048-9] [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: 07/02/2020] [Accepted: 02/08/2021] [Indexed: 10/22/2022]
Abstract
MiR-133b is considered to be lowly expressed in osteoporosis patients. This study aimed to probe the role and in-depth mechanism of miR-133b in modulating osteoblast biological behavior and differentiation. The differential expressions of miR-133b and GNB4 in patients with osteoporosis and healthy control were analyzed based on the GEO database. Osteoblastic differentiation of hFOB 1.19 cells was induced in the culture medium containing 10 mM β-glycerophosphate, 50 nm dexamethasone, and 100 μg/ml ascorbic acid. The level of GNB4 was detected using quantitative real-time PCR (qRT-PCR) and Western blot. Cell viability and apoptosis were measured by Cell Counting Kit-8 (CCK-8) and flow cytometry assays, respectively. Western blot was also utilized to measure the levels of osteoblast-related proteins, including ALP, Runx2, Osterix, and OPN. GNB4 was identified and confirmed as a downstream target gene of miR-133b. The expression of miR-133b was declined while the expression of GNB4 was increased in osteoporosis patients. Importantly, up-regulation of miR-133b caused the increase of cell viability and the decrease of apoptosis, which could be blocked by overexpression of GNB4. Also, up-regulation of miR-133b promoted osteoblasts differentiation, as shown by the increase in the expression of ALP, Runx2, Osterix, and OPN. Similarly, this promoting impact resulted from miR-133b overexpression can be reversed via up-regulation of GNB4. These findings revealed that miR-133b can promote the viability and differentiation of osteoblasts by targeting GNB4, hoping to lay a feasible theoretical foundation for the clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Jinqiang Wang
- Department of Spine Surgery, Weifang Traditional Chinese Medicine Hospital, No. 1055 Weizhou Road, Kuiwen District, Weifang, Shandong, People's Republic of China.
| | - Zhaoqing Gao
- Department of Spine Surgery, Weifang Traditional Chinese Medicine Hospital, No. 1055 Weizhou Road, Kuiwen District, Weifang, Shandong, People's Republic of China
| | - Peng Gao
- Department of Spine Surgery, Weifang Traditional Chinese Medicine Hospital, No. 1055 Weizhou Road, Kuiwen District, Weifang, Shandong, People's Republic of China
| |
Collapse
|
28
|
Nguyen DND, Chilian WM, Zain SM, Daud MF, Pung YF. MicroRNA regulation of vascular smooth muscle cells and its significance in cardiovascular diseases. Can J Physiol Pharmacol 2021; 99:827-838. [PMID: 33529092 DOI: 10.1139/cjpp-2020-0581] [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] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is among the leading causes of death worldwide. MicroRNAs (miRNAs), regulatory molecules that repress protein expression, have attracted considerable attention in CVD research. The vasculature plays a big role in CVD development and progression and dysregulation of vascular cells underlies the root of many vascular diseases. This review provides a brief introduction of the biogenesis of miRNAs and exosomes, followed by overview of the regulatory mechanisms of miRNAs in vascular smooth muscle cells (VSMCs) intracellular signaling during phenotypic switching, senescence, calcification, and neointimal hyperplasia. Evidence of extracellular signaling of VSMCs and other cells via exosomal and circulating miRNAs is also presented. Lastly, current drawbacks and limitations of miRNA studies in CVD research and potential ways to overcome these disadvantages are discussed in detail. In-depth understanding of VSMC regulation via miRNAs will add substantial knowledge and advance research in diagnosis, disease progression, and (or) miRNA-derived therapeutic approaches in CVD research.
Collapse
Affiliation(s)
- Duong Ngoc Diem Nguyen
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Semenyih, 43500 Selangor, Malaysia
| | - William M Chilian
- Integrative Medical Sciences, Northeast Ohio Medical University, 4209 St. Rt. 44, P.O. Box 95, Rootstown, OH P.O. Box 95, USA
| | - Shamsul Mohd Zain
- The Pharmacogenomics Laboratory, Department of Pharmacology, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Muhammad Fauzi Daud
- Institute of Medical Science Technology, Universiti Kuala Lumpur, Kajang, 43000 Selangor, Malaysia
| | - Yuh-Fen Pung
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Semenyih, 43500 Selangor, Malaysia
| |
Collapse
|
29
|
Choe N, Kwon DH, Ryu J, Shin S, Cho HJ, Joung H, Eom GH, Ahn Y, Park WJ, Nam KI, Kim YK, Kook H. miR-27a-3p Targets ATF3 to Reduce Calcium Deposition in Vascular Smooth Muscle Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:627-639. [PMID: 33230462 PMCID: PMC7578555 DOI: 10.1016/j.omtn.2020.09.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/23/2020] [Indexed: 01/16/2023]
Abstract
Vascular calcification, the ectopic deposition of calcium in blood vessels, develops in association with various metabolic diseases and atherosclerosis and is an independent predictor of morbidity and mortality associated with these diseases. Herein, we report that reduction of microRNA-27a-3p (miR-27a-3p) causes an increase in activating transcription factor 3 (ATF3), a novel osteogenic transcription factor, in vascular smooth muscle cells. Both microRNA (miRNA) and mRNA microarrays were performed with rat vascular smooth muscle cells, and reciprocally regulated pairs of miRNA and mRNA were selected after bioinformatics analysis. Inorganic phosphate significantly reduced the expression of miR-27a-3p in A10 cells. The transcript level was also reduced in vitamin D3-administered mouse aortas. miR-27a-3p mimic reduced calcium deposition, whereas miR-27a-3p inhibitor increased it. The Atf3 mRNA level was upregulated in a cellular vascular calcification model, and miR-27a-3p reduced the Atf3 mRNA and protein levels. Transfection with Atf3 could recover the miR-27a-3p-induced reduction of calcium deposition. Our results suggest that reduction of miR-27a-3p may contribute to the development of vascular calcification by de-repression of ATF3.
Collapse
Affiliation(s)
- Nakwon Choe
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Duk-Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Juhee Ryu
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea.,Department of Biochemistry, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Sera Shin
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Hye Jung Cho
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Hosouk Joung
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Gwang Hyeon Eom
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Kwang-Il Nam
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Hwasun, Jeollanamdo, Republic of Korea
| |
Collapse
|
30
|
Wang SS, Wang C, Chen H. MicroRNAs are critical in regulating smooth muscle cell mineralization and apoptosis during vascular calcification. J Cell Mol Med 2020; 24:13564-13572. [PMID: 33089928 PMCID: PMC7754013 DOI: 10.1111/jcmm.16005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 02/01/2023] Open
Abstract
Vascular calcification refers to the pathological deposition of calcium and phosphate minerals into the vasculature. It is prevalent in atherosclerosis, ageing, type 2 diabetes mellitus and chronic kidney disease, thus, increasing morbidity and mortality from these conditions. Vascular calcification shares similar mechanisms with bone mineralization, with smooth muscle cells playing a critical role in both processes. In the last decade, a variety of microRNAs have been identified as key regulators for the differentiation, phenotypic switch, proliferation, apoptosis, cytokine production and matrix deposition in vascular smooth muscle cells during vascular calcification. Therefore, this review mainly discusses the roles of microRNAs in the pathophysiological mechanisms of vascular calcification in smooth muscle cells and describes several interventions against vascular calcification by regulating microRNAs. As the exact mechanisms of calcification remain not fully elucidated, having a better understanding of microRNA involvement in vascular calcification may give impetus to development of novel therapeutics for the control and treatment of vascular calcification.
Collapse
Affiliation(s)
- Shan-Shan Wang
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Wang
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Han Chen
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
31
|
Duan M, Zhao WL, Zhou L, Novák P, Zhu X, Yin K. Omics research in vascular calcification. Clin Chim Acta 2020; 511:319-328. [PMID: 33096035 DOI: 10.1016/j.cca.2020.10.022] [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] [Received: 08/31/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC), the pathological process of hydroxyapatite mineral deposition in the vascular system, is closely associated with aging, atherosclerotic plaque formation, cardiovascular disease (CVD) and diabetes mellitus (DM). Studies have shown that VC is related to cellular phenotypic changes, extracellular vesicles, disordered calcium and phosphate homeostasis, and an imbalance between inducers and inhibitors of VC. Unfortunately, there is currently no effective preventive or targeted treatment for pathologic condition. The rapid evolution of omics technology (genomics, epigenomics, transcriptomics, proteomics and metabolomics) has provided a novel approach for elucidation of pathophysiologic mechanisms in general and those associated with VC specifically. Here, we review articles published over the last twenty years and focus on the current state, challenges, limitations and future of omics in VC research and clinical practice. Highlighting potential targets based on omics technology will improve our understanding of this pathologic condition and assist in the development of potential treatment options for VC related disease.
Collapse
Affiliation(s)
- Meng Duan
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Wen-Li Zhao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| |
Collapse
|
32
|
Duan M, Zhao WL, Zhou L, Novák P, Zhu X, Yin K. Omics research in vascular calcification. Clin Chim Acta 2020; 511:198-207. [PMID: 33096032 DOI: 10.1016/j.cca.2020.10.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
Vascular calcification (VC), the pathological process of hydroxyapatite mineral deposition in the vascular system, is closely associated with aging, atherosclerotic plaque formation, cardiovascular disease (CVD) and diabetes mellitus (DM). Studies have shown that VC is related to cellular phenotypic changes, extracellular vesicles, disordered calcium phosphate homeostasis and an imbalance between inducers and inhibitors of VC. Unfortunately, there is currently no effective preventive or targeted treatment for this disorder. Recently, the evolution of omics technology (genomics, epigenomics, transcriptomics, proteomics and metabolomics) has paved the way for elucidation of complex biochemical processes and, as such, may provide new insight on VC. Accordingly, we conducted a review of articles published over the last twenty years and herein focus on current and future potential of omics technology in clarifying mechanisms of this disease process. Identification of new biomarkers will provide additional tools in characterizing this pathology and will further assist in the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Meng Duan
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Wen-Li Zhao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| |
Collapse
|
33
|
Jiao J, Feng G, Wu M, Wang Y, Li R, Liu J. MiR-140-5p promotes osteogenic differentiation of mouse embryonic bone marrow mesenchymal stem cells and post-fracture healing of mice. Cell Biochem Funct 2020; 38:1152-1160. [PMID: 33047358 PMCID: PMC7756898 DOI: 10.1002/cbf.3585] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 07/09/2020] [Accepted: 08/01/2020] [Indexed: 12/13/2022]
Abstract
MiR-140-5p is high expressed in normal fracture healing, but its specific role and mechanism in tissue-to-bone healing are rarely reported. Therefore, this study investigated the effects of miR-140-5p on tissue-to-bone healing. Clone formation experiment, flow cytometry, Alizarin Red S Staining and Oil Red O Staining were performed to investigate the biological characteristics of mouse embryonic bone marrow mesenchymal stem cells C3H10T1/2. MiR-140-5p mimic was transfected into osteogenic medium (OS)-treated C3H10T1/2 cells to investigate the effects of miR-140-5p on osteogenic differentiation. MiR-140-5p transgenic mouse model and the transgenic fracture model were established, and the effects of miR-140-5p on osteogenic differentiation, bone mineral density (BMD) and bone mass of bone tissues were detected by haematoxylin and eosin staining and computed tomography scan. The expressions of osteocalcin, differentiation-related genes (Runx2, ALP, Spp1 and Bglap3) and miR-140-5p were determined by quantitative real-time polymerase chain reaction. C3H10T1/2 cells showed the abilities of forming cloned differentiation of osteogenesis, fat cells, and its phenotypes including CD44, CD90.1 and Sca-1 but excluding CD45 haematopoietic stem cell marker. Overexpression of miR-140-5p promoted the expressions of differentiation-related genes and calcium deposition of OS-treated C3H10T1/2 cells. MiR-140-5p increased the expression of osteocalcin, BMD and bone mass and promoted bone healing of miR-140-5p-transgenic mice with fracture. MiR-140-5p promoted osteogenic differentiation of mouse embryonic bone marrow mesenchymal stem cells and post-fracture healing in mice. SIGNIFICANCE OF THE STUDY: C3H10T1/2 cells showed the abilities of forming cloned differentiation of osteogenesis, fat cells and its phenotypes including CD44, CD90.1 and Sca-1 but excluding CD45 haematopoietic stem cell marker. Overexpression of miR-140-5p promoted the expressions of differentiation-related genes and calcium deposition of osteogenic medium-treated C3H10T1/2 cells. MiR-140-5p increased the expression of osteocalcin and bone mineral density and bone mass and promoted bone healing of miR-140-5p-transgenic mice with fracture. Our results showed that miR-140-5p promoted osteogenic differentiation of mouse embryonic bone marrow mesenchymal stem cells and post-fracture healing in mice, which may be a therapeutic target for treating fractures and promoting bone healing.
Collapse
Affiliation(s)
- Jianhang Jiao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Guang Feng
- The Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Minfei Wu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Yang Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jun Liu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
34
|
Ryu J, Ahn Y, Kook H, Kim YK. The roles of non-coding RNAs in vascular calcification and opportunities as therapeutic targets. Pharmacol Ther 2020; 218:107675. [PMID: 32910935 DOI: 10.1016/j.pharmthera.2020.107675] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is characterized by an accumulation of calcium phosphate crystals inside the vessel wall. VC is often associated with diabetes, chronic kidney disease (CKD), atherosclerosis, and cardiovascular disease (CVD). Even though the number of patients with VC remains prevalent, there are still no approved therapies for the treatment of VC. Since the pathogenesis of VC is diverse and involves multiple factors and mechanisms, it is critical to reveal the novel mechanisms involved in VC. Although protein-coding RNAs involved in VC have been extensively studied, the roles of non-coding RNAs (ncRNAs) are not yet fully understood. The field of ncRNAs has recently received attention, and accumulating evidence from studies in VC suggests that ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), play an important role in the regulation of VC. NcRNAs can modulate VC by acting as promoters or inhibitors and may be useful in the clinical diagnosis and treatment of VC. In this article, we review and discuss ncRNAs that regulate VC and present the therapeutic implications of these ncRNAs.
Collapse
Affiliation(s)
- Juhee Ryu
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Jeollanam-do, Republic of Korea; Department of Biochemistry, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Pharmacology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea
| | - Youngkeun Ahn
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Cardiology, Cardiovascular Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyun Kook
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Jeollanam-do, Republic of Korea; Department of Pharmacology, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.
| | - Young-Kook Kim
- Basic Research Laboratory for Vascular Remodeling, Chonnam National University Medical School, Jeollanam-do, Republic of Korea; Department of Biomedical Sciences, Center for Creative Biomedical Scientists at Chonnam National University, Jeollanam-do, Republic of Korea; Department of Biochemistry, Chonnam National University Medical School, Jeollanam-do, Republic of Korea.
| |
Collapse
|
35
|
Fang M, Liu K, Li X, Wang Y, Li W, Li B. AntagomiR-29b inhibits vascular and valvular calcification and improves heart function in rats. J Cell Mol Med 2020; 24:11546-11557. [PMID: 32845082 PMCID: PMC7576293 DOI: 10.1111/jcmm.15770] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 01/08/2023] Open
Abstract
We aimed to investigate the role of the miR‐29b and its effect on TGF‐β3 pathway in vascular and valvular calcification in a rat model of calcific aortic valve diseases (CAVD). A rat model of CAVD was established by administration of warfarin plus vitamin K. The expression levels of miR‐29b, osteogenic markers and other genes were determined by qRT‐PCR, Western blot and/or immunofluorescence and immunohistochemistry. The calcium content and alkaline phosphatase (ALP) activity were measured. The calcium content, ALP activity and osteogenic markers levels in calcified aorta and aortic valve were augmented compared to controls. The expression of miR‐29b, p‐Smad3, and Wnt3 and β‐catenin was significantly up‐regulated, whereas TGF‐β3 was markedly down‐regulated. However, compared with the CAVD model group, the calcium content and ALP activity in rats treated with antagomiR‐29b were significantly decreased, and antagomiR‐29b administration reversed the effects of CAVD model on the expression of miR‐29b and osteogenic markers. Inhibition of miR‐29b in CAVD rats prevented from vascular and valvular calcification and induced TGF‐β3 expression, suggesting that the miR‐29b/TGF‐β3 axis may play a regulatory role in the pathogenesis of vascular and valvular calcification and could play a significant role in the treatment of CAVD and other cardiovascular diseases.
Collapse
Affiliation(s)
- Ming Fang
- Department of Cardiology, Hainan General Hospital, Haikou, China.,Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Kangyong Liu
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xinming Li
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yudai Wang
- Department of Cardiology, Hainan General Hospital, Haikou, China
| | - Wei Li
- Department of Radiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Bin Li
- Department of Cardiology, Hainan General Hospital, Haikou, China
| |
Collapse
|
36
|
Zhang X, Chen J, Meng Q, Li D, Hu FZ, Zhu YQ, Huang YY, Liu YN, Sun L, Liang QH. The protective effects of long non-coding RNA-ANCR on arterial calcification. J Bone Miner Metab 2020; 38:421-431. [PMID: 31974677 DOI: 10.1007/s00774-019-01076-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/17/2019] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Arterial calcification is a major factor for cardiovascular events and is characterized by vascular smooth muscle cells (VSMCs) transformed into osteoblast-like cells. Long non-coding RNAs (lncRNA) were recognized as important regulators of diverse biological processes. Previous studies have demonstrated that lncRNAs could regulate the proliferation and apoptosis of VSMCs. LncRNA-ANCR (Anti-differentiation ncRNA) is an essential mediator governing the differentiation of human osteoblast. However, it is unclear whether ANCR could regulate the osteoblastic differentiation of VSMCs. In this study, we determined the effect of ANCR on VSMCs differentiation and arterial calcification. MATERIALS AND METHODS Both cellular and mouse model of arterial calcification were, respectively, established to investigate the role of ANCR in the mechanism of arterial calcification. ANCR overexpressing lentivirus were used to investigate the effects of ANCR on the expression of bone proteins and autophagy-related molecules. RESULTS ANCR could inhibit β-glycerophosphate (β-GP)-induced VSMCs osteoblastic differentiation and mineralization due to decreased expressions of Runt-related transcription factor 2, bone morphogenetic protein-2, and formation of mineralized nodule, and attenuate high calcitriol-induced mice model of arterial calcification. Furthermore, ANCR could significantly increase LC3 and autophagy protein 5 expression in β-GP-stimulated VSMCs, and the effect could be inhibited by 3-methyladenine, a pharmacological inhibitor of autophagy. CONCLUSION ANCR may inhibit the osteoblastic differentiation of VSMCs and attenuate mice arterial calcification through activating autophagy.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Jing Chen
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Qiang Meng
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Dong Li
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Fang-Zhi Hu
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Yu-Qing Zhu
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Yuan-Yuan Huang
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Ya-Nan Liu
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China
| | - Lin Sun
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China.
| | - Qiu-Hua Liang
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People's Republic of China.
| |
Collapse
|
37
|
Tang X, Bai Y, Zhang Z, Lu J. A validated miRNA signature for the diagnosis of osteoporosis related fractures using SVM algorithm classification. Exp Ther Med 2020; 20:2209-2217. [PMID: 32765697 PMCID: PMC7401749 DOI: 10.3892/etm.2020.8928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/29/2020] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to develop a circulating microRNA expression signature for early prediction of osteoporotic fractures and to validate the results using Gene Expression Omnibus (GEO) datasets. The GSE70318 dataset was downloaded from GEO and used to build an osteoporotic fracture prediction model based on the receiver operating characteristic curve and support vector machine (SVM) classification index. The GSE74209 dataset was used as a validation dataset. Additionally, in vitro, alkaline phosphatase (ALP) activity was measured in the presence or absence of microRNA (miRNA/miR) treatments in human osteoblast cells. The expression of two selected genes was detected by western blotting. miR-188-3p, miR-942-3p, miR-576-3p and miR-135a-5p were differentially expressed between controls and osteoporotic patients with fractures. SVM classification using these four miRNAs provided better dichotomization. It was further confirmed that miR-576-3p and 135a-5p in the GSE74209 dataset could also significantly discriminate between the controls and fracture patients, the area under the curve of SVM2 was 0.9722 with 95% CI 0.8885-1.056. Further analysis indicated that the target genes of the two miRNAs participated in the Wingless-related integration site, Hedgehog and transforming growth factor-β signaling pathways and osteoclast differentiation. miR-576-3p and miR-135-5p transfection decreased ALP activity and ALP activity was increased in the presence of blocking antisense oligonucleotides. Western blotting indicated miR-576-3p and miR-135-5p decreased CSNK1A1L and LRP6 levels, respectively. In conclusion, two miRNA signatures were developed and validated for the prediction of osteoporotic fractures.
Collapse
Affiliation(s)
- Xiaolin Tang
- Department of Medical Science, Shunde Polytechnic, Foshan, Guangdong 528300, P.R. China
| | - Yinshan Bai
- Life Science and Engineering College, Foshan University, Foshan, Guangdong 528231, P.R. China
| | - Zhiming Zhang
- Department of Medical Science, Shunde Polytechnic, Foshan, Guangdong 528300, P.R. China
| | - Jianlin Lu
- Department of Medical Science, Shunde Polytechnic, Foshan, Guangdong 528300, P.R. China
| |
Collapse
|
38
|
Zununi Vahed S, Mostafavi S, Hosseiniyan Khatibi SM, Shoja MM, Ardalan M. Vascular Calcification: An Important Understanding in Nephrology. Vasc Health Risk Manag 2020; 16:167-180. [PMID: 32494148 PMCID: PMC7229867 DOI: 10.2147/vhrm.s242685] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular calcification (VC) is a life-threatening state in chronic kidney disease (CKD). High cardiovascular mortality and morbidity of CKD cases may root from medial VC promoted by hyperphosphatemia. Vascular calcification is an active, highly regulated, and complex biological process that is mediated by genetics, epigenetics, dysregulated form of matrix mineral metabolism, hormones, and the activation of cellular signaling pathways. Moreover, gut microbiome as a source of uremic toxins (eg, phosphate, advanced glycation end products and indoxyl-sulfate) can be regarded as a potential contributor to VC in CKD. Here, an update on different cellular and molecular processes involved in VC in CKD is discussed to elucidate the probable therapeutic pathways in the future.
Collapse
Affiliation(s)
| | - Soroush Mostafavi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammadali M Shoja
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | | |
Collapse
|
39
|
Roles of Histone Acetylation Modifiers and Other Epigenetic Regulators in Vascular Calcification. Int J Mol Sci 2020; 21:ijms21093246. [PMID: 32375326 PMCID: PMC7247359 DOI: 10.3390/ijms21093246] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/28/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification (VC) is characterized by calcium deposition inside arteries and is closely associated with the morbidity and mortality of atherosclerosis, chronic kidney disease, diabetes, and other cardiovascular diseases (CVDs). VC is now widely known to be an active process occurring in vascular smooth muscle cells (VSMCs) involving multiple mechanisms and factors. These mechanisms share features with the process of bone formation, since the phenotype switching from the contractile to the osteochondrogenic phenotype also occurs in VSMCs during VC. In addition, VC can be regulated by epigenetic factors, including DNA methylation, histone modification, and noncoding RNAs. Although VC is commonly observed in patients with chronic kidney disease and CVD, specific drugs for VC have not been developed. Thus, discovering novel therapeutic targets may be necessary. In this review, we summarize the current experimental evidence regarding the role of epigenetic regulators including histone deacetylases and propose the therapeutic implication of these regulators in the treatment of VC.
Collapse
|
40
|
Xu F, Zhong J, Lin X, Shan S, Guo B, Zheng M, Wang Y, Li F, Cui R, Wu F, Zhou E, Liao X, Liu Y, Yuan L. Melatonin alleviates vascular calcification and ageing through exosomal miR-204/miR-211 cluster in a paracrine manner. J Pineal Res 2020; 68:e12631. [PMID: 31943334 PMCID: PMC7154654 DOI: 10.1111/jpi.12631] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 12/08/2019] [Accepted: 01/06/2020] [Indexed: 12/24/2022]
Abstract
In the elderly with atherosclerosis, hypertension and diabetes, vascular calcification and ageing are ubiquitous. Melatonin (MT) has been demonstrated to impact the cardiovascular system. In this study, we have shown that MT alleviates vascular calcification and ageing, and the underlying mechanism involved. We found that both osteogenic differentiation and senescence of vascular smooth muscle cells (VSMCs) were attenuated by MT in a MT membrane receptor-dependent manner. Moreover, exosomes isolated from VSMCs or calcifying vascular smooth muscle cells (CVSMCs) treated with MT could be uptaken by VSMCs and attenuated the osteogenic differentiation and senescence of VSMCs or CVSMCs, respectively. Moreover, we used conditional medium from MT-treated VSMCs and Transwell assay to confirm exosomes secreted by MT-treated VSMCs attenuated the osteogenic differentiation and senescence of VSMCs through paracrine mechanism. We also found exosomal miR-204/miR-211 mediated the paracrine effect of exosomes secreted by VSMCs. A potential target of these two miRs was revealed to be BMP2. Furthermore, treatment of MT alleviated vascular calcification and ageing in 5/6-nephrectomy plus high-phosphate diet-treated (5/6 NTP) mice, while these effects were partially reversed by GW4869. Exosomes derived from MT-treated VSMCs were internalised into mouse artery detected by in vivo fluorescence image, and these exosomes reduced vascular calcification and ageing of 5/6 NTP mice, but both effects were largely abolished by inhibition of exosomal miR-204 or miR-211. In summary, our present study revealed that exosomes from MT-treated VSMCs could attenuate vascular calcification and ageing in a paracrine manner through an exosomal miR-204/miR-211.
Collapse
Affiliation(s)
- Feng Xu
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Jia‐Yu Zhong
- Department of GeriatricsInstitute of Aging and Age‐related Disease ResearchThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Xiao Lin
- Department of RadiologyThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Su‐Kang Shan
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Bei Guo
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Ming‐Hui Zheng
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Yi Wang
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Fuxingzi Li
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Rong‐Rong Cui
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Feng Wu
- Department of PathologyThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - En Zhou
- Department of Otorhinolaryngology Head and Neck SurgeryHunan Provincial People's HospitalChangshaChina
| | - Xiao‐Bo Liao
- Department of Cardiovascular SurgeryThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - You‐Shuo Liu
- Department of GeriatricsInstitute of Aging and Age‐related Disease ResearchThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Ling‐Qing Yuan
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
41
|
Cheng S, Zhu CH, Zhang AH, Huang SM. MiR-29b expression is altered in crescent formation of HSPN and accelerates Ang II-induced mesangial cell activation. World J Pediatr 2020; 16:201-212. [PMID: 31630337 DOI: 10.1007/s12519-019-00318-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/24/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND MicroRNA-29b (miR-29b) has been suggested to possess pro-inflammatory activity, which can partially be explained by the repression of tumor necrosis factor alpha protein three antibody (TNFAIP3). Meanwhile, it also promotes thyroid cell proliferation via Smad signaling pathways. The present study aimed to elucidate the role of miR-29b in Henoch Schönlein purpura nephritis (HSPN) and its underlying molecular mechanism in angiotensin II (Ang II)-induced human glomerular mesangial cell (HGMC) activation. METHODS We evaluated miR-29b expression in 35 HSPN renal tissues based on crescent formation, glomerular sclerosis, interstitial fibrosis, thrombosis formation and capillary loop necrosis. Meanwhile, HGMCs were cultured, treated with Ang II and then transfected with LV-hsa-miR-29b-1 to induce miR-29b overexpression or LV-hsa-miR-29b-3p-inhibition to inhibit miR-29b expression. Finally, we examined the effects of miR-29b on cell proliferation and release of inflammatory mediators. RESULTS We observed that miR-29b expression was significantly higher in the crescent group than in the no crescent group. MiR-29b overexpression induced the release of intercellular adhesion molecule-1, interleukin-1β (IL-1β), IL-6, IL-8, the increase of CyclinA2, CyclinD1, and cell proliferation. It also could inhibit the expressions of TNFAIP3 and NF-kappa-B-repressing factor (NKRF). Correspondingly, miR-29b inhibition produced the opposite effects and increased the expression of TNFAIP3 and NKRF. CONCLUSION MiR-29b expression is altered in crescent formation of HSPN and accelerates Ang II-induced mesangial cell proliferation and release of inflammatory mediators.
Collapse
Affiliation(s)
- Shan Cheng
- Department of Infectious Disease, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Chun-Hua Zhu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Ai-Hua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China
| | - Song-Ming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
| |
Collapse
|
42
|
The Epigenetic Landscape of Vascular Calcification: An Integrative Perspective. Int J Mol Sci 2020; 21:ijms21030980. [PMID: 32024140 PMCID: PMC7037112 DOI: 10.3390/ijms21030980] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/26/2022] Open
Abstract
Vascular calcification (VC) is an important complication among patients of advanced age, those with chronic kidney disease, and those with diabetes mellitus. The pathophysiology of VC encompasses passive occurrence of physico-chemical calcium deposition, active cellular secretion of osteoid matrix upon exposure to metabolically noxious stimuli, or a variable combination of both processes. Epigenetic alterations have been shown to participate in this complex environment, through mechanisms including DNA methylation, non-coding RNAs, histone modifications, and chromatin changes. Despite such importance, existing reviews fail to provide a comprehensive view of all relevant reports addressing epigenetic processes in VC, and cross-talk between different epigenetic machineries is rarely examined. We conducted a systematic review based on PUBMED and MEDLINE databases up to 30 September 2019, to identify clinical, translational, and experimental reports addressing epigenetic processes in VC; we retrieved 66 original studies, among which 60.6% looked into the pathogenic role of non-coding RNA, followed by DNA methylation (12.1%), histone modification (9.1%), and chromatin changes (4.5%). Nine (13.6%) reports examined the discrepancy of epigenetic signatures between subjects or tissues with and without VC, supporting their applicability as biomarkers. Assisted by bioinformatic analyses blending in each epigenetic component, we discovered prominent interactions between microRNAs, DNA methylation, and histone modification regarding potential influences on VC risk.
Collapse
|
43
|
Maternal Obesity in Mice Exacerbates the Allergic Inflammatory Response in the Airways of Male Offspring. Nutrients 2019; 11:nu11122902. [PMID: 31805682 PMCID: PMC6950392 DOI: 10.3390/nu11122902] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/11/2019] [Accepted: 11/19/2019] [Indexed: 02/05/2023] Open
Abstract
It was previously demonstrated that non-allergen-sensitized rodents born to mothers exposed to a high-fat diet (HFD) spontaneously develop lower respiratory compliance and higher respiratory resistance. In the present study, we sought to determine if mice born to mothers consuming HFD would exhibit changes in inflammatory response and lung remodeling when subjected to ovalbumin (OVA) sensitization/challenge in adult life. Mice born to dams consuming either HFD or standard chow had increased bronchoalveolar lavage (BAL) levels of IL-1β, IL-4, IL-5, IL-10, IL-13, TNF-α and TGF-β1 after challenge with OVA. IL-4, IL-13, TNF-α and TGF-β1 levels were further increased in the offspring of HFD-fed mothers. Mice born to obese dams also had exacerbated values of leukocyte infiltration in lung parenchyma, eosinophil and neutrophil counts in BAL, mucus overproduction and collagen deposition. The programming induced by maternal obesity was accompanied by increased expression of miR-155 in peripheral-blood mononuclear cells and reduced miR-133b in trachea and lung tissue in adult life. Altogether, the present data support the unprecedented notion that the progeny of obese mice display exacerbated responses to sensitization/challenge with OVA, leading to the intensification of the morphological changes of lung remodeling. Such changes are likely to result from long-lasting changes in miR-155 and miR-133b expression.
Collapse
|
44
|
The relation between serum phosphorus levels and long-term mortality in Chinese patients with ST-segment elevation myocardial infarction. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2019; 16:775-781. [PMID: 31700517 PMCID: PMC6828604 DOI: 10.11909/j.issn.1671-5411.2019.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Elevated serum phosphorus levels may be associated with adverse outcomes in cardiovascular disease. This study aimed to investigate the relation between serum phosphorus levels and risk of all-cause mortality in Chinese patients with ST-segment elevation myocardial infarction (STEMI) who had preserved renal function at baseline. Methods We enrolled patients with STEMI who had preserved renal function at baseline in Xuanwu Hospital from January 2011 to December 2016. Those patients were divided into four groups based on serum phosphorus levels. All-cause mortality rates were compared between groups. Mean duration of follow up was 54.6 months. We used Cox proportional-hazards models to examine the relation between serum phosphorus levels and all-cause mortality after adjustment for potential confounders. Results 1989 patients were involved and 211 patients (10.6%) died during follow-up. Based on serum phosphorus levels, patients were categorized into the following groups: < 2.50 mg/dL (n = 89), 2.51–3.50 mg/dL (n = 1066), 3.51–4.50 mg/dL (n = 672) and > 4.50 mg/dL (n = 162), respectively. The lowest mortality occurred in patients with serum phosphorus levels between 2.51–3.50 mg/dL, with a multivariable-adjusted hazard ratio of 1.19 (95% CI: 0.64–1.54), 1.37 (95% CI: 1.22–1.74), and 1.46 (95% CI: 1.35–1.83) in patients with serum phosphorus levels of < 2.50 mg/dL, 3.51–4.50 mg/dL and > 4.50 mg/dL, respectively. Conclusions Elevated serum phosphorus levels were associated with all-cause mortality in Chinese patients with STEMI who had preserved renal function at baseline.
Collapse
|
45
|
miR-149-3p Regulates the Switch between Adipogenic and Osteogenic Differentiation of BMSCs by Targeting FTO. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:590-600. [PMID: 31382190 PMCID: PMC6690430 DOI: 10.1016/j.omtn.2019.06.023] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/04/2019] [Accepted: 06/07/2019] [Indexed: 01/08/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) have been suggested to possess the capacity to differentiate into different cell lineages. Maintaining a balanced stem cell differentiation program is crucial to the bone microenvironment and bone development. MicroRNAs (miRNAs) have played a critical role in regulating the differentiation of BMSCs into particular lineage. However, the role of miR-149-3p in the adipogenic and osteogenic differentiation of BMSCs has not been extensively discovered. In this study, we aimed to detect the expression levels of miR-149-3p during the differentiation of BMSCs and investigate whether miR-149-3p participated in the lineage choice of BMSCs or not. Compared with mimic-negative control (NC), miR-149-3p mimic decreased the adipogenic differentiation potential of BMSCs and increased the osteogenic differentiation potential. Further analysis revealed that overexpression of miR-149-3p repressed the expression of fat mass and obesity-associated (FTO) gene through binding to the 3ʹ UTR of the FTO mRNA. Also, the role of miR-149-3p mimic in inhibiting adipogenic lineage differentiation and potentiating osteogenic lineage differentiation was mainly through targeting FTO, which also played an important role in regulating body weight and fat mass. In addition, BMSCs treated with miR-149-3p anti-miRNA oligonucleotide (AMO) exhibited higher potential to differentiate into adipocytes and lower tendency to differentiate into osteoblasts compared with BMSCs transfected with NC. In summary, our results detected the effects of miR-149-3p in cell fate specification of BMSCs and revealed that miR-149-3p inhibited the adipogenic differentiation of BMSCs via a miR-149-3p/FTO regulatory axis. This study provided cellular and molecular insights into the observation that miR-149-3p was a prospective candidate gene for BMSC-based bone tissue engineering in treating osteoporosis.
Collapse
|
46
|
Wang C, Tang Y, Wang Y, Li G, Wang L, Li Y. Label-free quantitative proteomics identifies Smarca4 is involved in vascular calcification. Ren Fail 2019; 41:220-228. [PMID: 30973285 PMCID: PMC6461080 DOI: 10.1080/0886022x.2019.1591997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Vascular calcification (VC) is a pathological process characterized by abnormal deposition of calcium phosphate, hydroxyapatite and other mineral substances in the vascular wall. Hyperphosphorus is an important risk factor associated with VC in the general population and patients with chronic kidney disease (CKD). However, there is still a lack of early biomarkers for hyperphosphorus induced VC. We established a calcific rat aorta vascular smooth muscle cells (RASMCs) model by stimulating with β-glycerophosphate. Then we performed label-free quantitative proteomics combined with liquid chromatograph–mass spectrometer/mass spectrometer (LC-2D-MS/MS)analysis and bioinformatics analysis to find the potential biomarkers for VC. In the current study, we identified 113 significantly proteins. Fifty six of these proteins were significantly up-regulated and the other 57 proteins were significantly decreased in calcific RASMCs, compared to that of normal control cells (fold-change (fc)>1.2, p < .05). Bioinformatics analysis indicated that these significant proteins mainly involved in the placenta blood vessel development and liver regeneration. Their molecule function was cell adhesion molecule binding. Among them, Smarca4 is significantly up-regulated in calcific RASMCs with fc = 2.72 and p = .01. In addition, we also established VC rat model. Real-time quantitative PCR analysis confirmed that the expression of Smarca4 was significantly increased in the aorta of calcified rat. Consistent with the up-regulation of Smarca4, the expression of VC associated microRNA such as miR-133b and miR-155 was also increased. Consequently, our study demonstrates that Smarca4 is involved in hyperphosphorus-induced VC. This finding may contribute to the early diagnosis and prevention of VC.
Collapse
Affiliation(s)
- Chan Wang
- a Department of Nephrology , Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China , Chengdu , China.,b Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan People's Hospital , Chengdu , China
| | - Yun Tang
- a Department of Nephrology , Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China , Chengdu , China.,b Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan People's Hospital , Chengdu , China
| | - Yanmei Wang
- c Department of Nephrology , Affiliated Hospital of North Sichuan Medical College , Nanchong , China
| | - Guisen Li
- a Department of Nephrology , Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China , Chengdu , China.,b Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan People's Hospital , Chengdu , China
| | - Li Wang
- a Department of Nephrology , Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China , Chengdu , China
| | - Yi Li
- a Department of Nephrology , Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China , Chengdu , China.,b Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan People's Hospital , Chengdu , China
| |
Collapse
|
47
|
Diverse roles of noncoding RNAs in vascular calcification. Arch Pharm Res 2019; 42:244-251. [PMID: 30673937 DOI: 10.1007/s12272-019-01118-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/15/2019] [Indexed: 12/19/2022]
Abstract
Vascular calcification occurs when calcium phosphate crystals are abnormally deposited in the vessel walls, thus hardening and narrowing the arteries. This condition is commonly observed in patients with diseases such as atherosclerosis, chronic kidney disease, diabetes, and cardiovascular diseases. Despite many studies being conducted, the molecular mechanism involved in vascular calcification is unknown. From recent studies, it is clear that several types of noncoding RNAs are involved in human diseases. It has also been shown that the noncoding RNAs, including microRNAs, long noncoding RNAs, and circular RNAs, are involved in the progression of vascular calcification. With the development of therapeutic approaches based on the manipulation of noncoding RNAs, it is speculated that the modulation of these molecules could be another strategy to treat vascular calcification in the future. In this review, we summarize the roles of various noncoding RNAs in vascular calcification and the technologies to modulate the noncoding RNAs for therapeutic purpose.
Collapse
|
48
|
Abbasian N, Herbert KE, Pawluczyk I, Burton JO, Bevington A. Vesicles bearing gifts: the functional importance of micro-RNA transfer in extracellular vesicles in chronic kidney disease. Am J Physiol Renal Physiol 2018; 315:F1430-F1443. [PMID: 30110570 DOI: 10.1152/ajprenal.00318.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), including microparticles (MPs) and exosomes (EXOs), are derived from a wide range of mammalian cells including blood platelets, endothelial cells, and kidney cells and can be detected in body fluids including blood and urine. While EVs are well established as diagnostic markers under pathophysiological and stress conditions, there is also mounting evidence of their functional significance as vehicles for communication between cells mediated by the presence of nucleic acids, especially microRNAs (miRs), encapsulated in the EVs. miRs regulate gene expression, are transported both in MPs and EXOs, and exert profound effects in the kidney. Here we review current understanding of the links between EVs and miRs, discuss the importance of miRs in kidney disease, and shed light on the role of EVs in transferring miRs through the circulation among the renal, vascular, and inflammatory cell populations that are functionally important in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Nima Abbasian
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - Karl E Herbert
- Department of Cardiovascular Sciences, University of Leicester, and Leicester National Institute of Health Research Cardiovascular Biomedical Research Unit , Leicester , United Kingdom
| | - Izabella Pawluczyk
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - James O Burton
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom.,John Walls Renal Unit, University Hospitals of Leicester , Leicester , United Kingdom
| | - Alan Bevington
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| |
Collapse
|
49
|
Lin X, Xu F, Cui RR, Xiong D, Zhong JY, Zhu T, Li F, Wu F, Xie XB, Mao MZ, Liao XB, Yuan LQ. Arterial Calcification Is Regulated Via an miR-204/DNMT3a Regulatory Circuit Both In Vitro and in Female Mice. Endocrinology 2018; 159:2905-2916. [PMID: 29850805 DOI: 10.1210/en.2018-00320] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/24/2018] [Indexed: 12/16/2022]
Abstract
Arterial calcification is a common cardiovascular disease that initiates from a process of osteoblastic differentiation of vascular smooth muscle cells (VSMCs). Accumulating evidence has demonstrated that microRNAs play an important role in regulating arterial calcification. miR-204 was significantly downregulated in calcified human renal arteries from patients with uremia; calcified arteries of mice, due to 5/6 nephrectomy with a high-phosphate diet (5/6 NTP); and in VSMCs induced by high phosphate concentration. The overexpression of miR-204 alleviated the osteoblastic differentiation of VSMCs. Bisulphite sequencing PCR revealed that CpG sites upstream of miR-204 DNA were hypermethylated in calcified VSMCs; in calcified arteries of mice, due to 5/6 NTP; and in calcified renal artery tissues from patients with uremia. Moreover, increased DNMT3a resulted in the hypermethylation of miR-204 in high phosphate concentration-induced VSMCs, whereas 5-aza-2'-deoxycytidine could restore the expression of miR-204 in high phosphate concentration-induced VSMCs. Moreover, we found that DNMT3a was the target of miR-204, and the methylation ratio of miR-204 was decreased significantly, meaning that the expression of miR-204 was restored when DNMT3a was knocked down by using DNMT3a small interfering RNA, resulting in abrogation of the effect of high phosphate concentration on VSMC calcification. The progress of arterial calcification is regulated by the miR-204/DNMT3a regulatory circuit.
Collapse
Affiliation(s)
- Xiao Lin
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Feng Xu
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Rong-Rong Cui
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Dan Xiong
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jia-Yu Zhong
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Ting Zhu
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Fuxingzi Li
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Feng Wu
- Department of Pathology, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xu-Biao Xie
- Center of Organ Transplantation, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Min-Zhi Mao
- Department of Orthopedics, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xiao-Bo Liao
- Department of Cardiovascular Surgery, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Ling-Qing Yuan
- Department of Endocrinology and Metabolism, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
50
|
Fang M, Wang C, Zheng C, Luo J, Hou S, Liu K, Li X. Mir-29b promotes human aortic valve interstitial cell calcification via inhibiting TGF-β3 through activation of wnt3/β-catenin/Smad3 signaling. J Cell Biochem 2018; 119:5175-5185. [PMID: 29227539 PMCID: PMC6001435 DOI: 10.1002/jcb.26545] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/30/2017] [Indexed: 01/08/2023]
Abstract
Herein, we hypothesized that pro-osteogenic MicroRNAs (miRs) could play functional roles in the calcification of the aortic valve and aimed to explore the functional role of miR-29b in the osteoblastic differentiation of human aortic valve interstitial cells (hAVICs) and the underlying molecular mechanism. Osteoblastic differentiation of hAVICs isolated from human calcific aortic valve leaflets obtained intraoperatively was induced with an osteogenic medium. Alizarin red S staining was used to evaluate calcium deposition. The protein levels of osteogenic markers and other proteins were evaluated using western blotting and/or immunofluorescence while qRT-PCR was applied for miR and mRNA determination. Bioinformatics and luciferase reporter assay were used to identify the possible interaction between miR-29b and TGF-β3. Calcium deposition and the number of calcification nodules were pointedly and progressively increased in hAVICs during osteogenic differentiation. The levels of osteogenic and calcification markers were equally increased, thus confirming the mineralization of hAVICs. The expression of miR-29b was significantly increased during osteoblastic differentiation. Furthermore, the osteoblastic differentiation of hAVICs was significantly inhibited by the miR-29b inhibition. TGF-β3 was markedly downregulated while Smad3, Runx2, wnt3, and β-catenin were significantly upregulated during osteogenic induction at both the mRNA and protein levels. These effects were systematically induced by miR-29b overexpression while the inhibition of miR-29b showed the inverse trends. Moreover, TGF-β3 was a direct target of miR-29b. Inhibition of miR-29b hinders valvular calcification through the upregulation of the TGF-β3 via inhibition of wnt/β-catenin and RUNX2/Smad3 signaling pathways.
Collapse
Affiliation(s)
- Ming Fang
- Department of CardiologyHainan General HospitalHaikouHainanP.R. China
- Department of CardiologyShanghai Zhoupu HospitalShanghaiP.R. China
| | - Cheng‐Guang Wang
- Laboratory of System BiologyShanghai Advanced Research InstituteChinese Academy of SciencesShanghaiP.R. China
| | - Changzhu Zheng
- Department of CardiologyShanghai Zhoupu HospitalShanghaiP.R. China
| | - Jun Luo
- Department of CardiologyShanghai Zhoupu HospitalShanghaiP.R. China
| | - Shiqiang Hou
- Department of CardiologyShanghai Zhoupu HospitalShanghaiP.R. China
| | - Kangyong Liu
- Department of NeurologyShanghai Zhoupu HospitalShanghaiP.R. China
| | - Xinming Li
- Department of CardiologyShanghai Zhoupu HospitalShanghaiP.R. China
| |
Collapse
|