1
|
Zimbru RI, Zimbru EL, Bojin FM, Haidar L, Andor M, Harich OO, Tănasie G, Tatu C, Mailat DE, Zbîrcea IM, Hirtie B, Uța C, Bănărescu CF, Panaitescu C. Connecting the Dots: How MicroRNAs Link Asthma and Atherosclerosis. Int J Mol Sci 2025; 26:3570. [PMID: 40332077 PMCID: PMC12026532 DOI: 10.3390/ijms26083570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Asthma and atherosclerosis are chronic conditions with distinct pathophysiologies, but overlapping inflammatory mechanisms that suggest a potential common regulatory framework. MicroRNAs (miRNAs), small non-coding RNA molecules that modulate gene expression post-transcriptionally, could be key players in linking these disorders. This review outlines how miRNAs contribute to the complex interplay between asthma and atherosclerosis, focusing on key miRNAs involved in inflammatory pathways, immune cell regulation and vascular remodeling. We discuss specific miRNAs, such as miR-155, miR-21 and miR-146a, which have been shown to modulate inflammatory cytokine production and T cell differentiation, impacting respiratory and cardiovascular health. The common miRNAs found in both asthma and atherosclerosis emphasize their role as potential biomarkers, but also as therapeutic targets. Understanding these molecular connections may unlock novel approaches for innovative, integrated treatment strategies that address both conditions and may significantly improve patient outcomes. Further research is needed to explore mechanistic pathways and validate the translational potential of miRNA-based interventions in preclinical and clinical settings.
Collapse
Affiliation(s)
- Răzvan-Ionuț Zimbru
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Elena-Larisa Zimbru
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Multidisciplinary Heart Research Center, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Florina-Maria Bojin
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Laura Haidar
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
| | - Minodora Andor
- Multidisciplinary Heart Research Center, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Cardiology Clinic, Timisoara Municipal Clinical Emergency Hospital, 12 Revoluției din 1989 Bd., 300040 Timisoara, Romania
| | - Octavia Oana Harich
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
| | - Gabriela Tănasie
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Carmen Tatu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Diana-Evelyne Mailat
- Multidisciplinary Heart Research Center, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Cardiology Clinic, Timisoara Municipal Clinical Emergency Hospital, 12 Revoluției din 1989 Bd., 300040 Timisoara, Romania
| | - Iulia-Maria Zbîrcea
- Department of Automation and Applied Informatics, “Politehnica” University of Timisoara, 300006 Timișoara, Romania
| | - Bogdan Hirtie
- ENT Department, “Victor Babes” University of Medicine and Pharmacy, 300042 Timișoara, Romania
| | - Cristina Uța
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
- Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Camelia-Felicia Bănărescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
- Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Carmen Panaitescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.-I.Z.)
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
- Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| |
Collapse
|
2
|
Rodelo-Haad C, Rodríguez-Ortiz ME, Garcia-Sáez R, Rivas-Domínguez A, Jurado-Montoya D, Martín-Malo A, Rodríguez M, Pendón-Ruiz de Mier MV, Muñoz-Castañeda JR. The true cost of phosphate control in chronic kidney disease. Clin Kidney J 2025; 18:i46-i60. [PMID: 40083951 PMCID: PMC11903093 DOI: 10.1093/ckj/sfae434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Indexed: 03/16/2025] Open
Abstract
The loss of kidney function entails the development of a positive phosphate balance. The burden of addressing elevated phosphate levels is high. Both parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) are increased to promote phosphaturia, thereby preventing the rise in serum phosphate. However, if the phosphate load is excessive, the corresponding phosphaturia is maximal, kidney function deteriorates and hyperphosphataemia becomes clinically evident in advanced stages of chronic kidney disease (CKD). In addition to its role in CKD progression, hyperphosphataemia has been linked to a multitude of adverse outcomes, including overt inflammation, vascular calcifications, endothelial dysfunction, cardiovascular disease, renal osteodystrophy and secondary hyperparathyroidism. Collectively, these factors contribute to the markedly elevated mortality rates observed among individuals with CKD. Furthermore, hyperphosphataemia has been identified as a significant contributor to the development of inflammatory processes, oxidative stress and fibrosis, which underlie the aetiology of numerous comorbidities. Additionally, elevated levels of PTH and FGF23 have been demonstrated to independently induce organ and tissue injury, which is associated with poor outcomes in CKD. This article provides a concise overview of the current understanding of phosphate handling by the kidney in the context of CKD. It outlines the detrimental effects of phosphate on various organs and the mechanisms through which it contributes to CKD progression. Additionally, we discuss the tools available for clinicians to identify patients at risk of an excessive phosphate load.
Collapse
Affiliation(s)
- Cristian Rodelo-Haad
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - María E Rodríguez-Ortiz
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - Raquel Garcia-Sáez
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Antonio Rivas-Domínguez
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Daniel Jurado-Montoya
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Alejandro Martín-Malo
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - Mariano Rodríguez
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
- European Uremic Toxins Group (EUTOx)
- COST Action CA21165 – Personalized medicine in chronic kidney disease: improved outcome based on Big Data (PerMediK)
| | - M Victoria Pendón-Ruiz de Mier
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - Juan Rafael Muñoz-Castañeda
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
3
|
Wang Z, Gui Z, Zhang L, Wang Z. Advances in the mechanisms of vascular calcification in chronic kidney disease. J Cell Physiol 2025; 240: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
|
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
|
5
|
Ye G, Liu T, Ding C. Bioinformatics analysis of key genes and potential therapeutic agents for vascular calcification in chronic kidney disease. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024:1-19. [PMID: 39556059 DOI: 10.1080/15257770.2024.2423214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 11/19/2024]
Abstract
Vascular calcification is a common complication of chronic kidney disease (CKD). The molecular mechanisms underlying this condition and the efficacy of potential treatments remain unclear. Bioinformatic methods were employed to analyze gene ontology (GO) annotations and pathway enrichments. Subsequently, an analysis of potential therapeutic agents for vascular calcification in CKD was performed. A total of 76 common genes, 181 enriched GO annotations-comprising 153 biological processes, 10 cellular components, and 18 molecular functions-41 KEGG pathways, 13 REACTOME pathways, and 3 BIOCARTA pathways were identified. Five key genes (PSMC5, TNFSF11, TNFRSF11A, TNFRSF12A, and ICAM1) were isolated. Most notably, the top five potential therapeutic drugs-ENAVATUZUMAB, DENOSUMAB, ALICAFORSEN, BI-505, and ENLIMOMAB PEGOL-were identified for vascular calcification in CKD. However, further molecular biological experiments are required to confirm these findings.
Collapse
Affiliation(s)
- Guojie Ye
- Cardiac Department, Aerospace Center Hospital, Beijing, China
- Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Tengfei Liu
- Cardiac Department, Aerospace Center Hospital, Beijing, China
- Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Chunhua Ding
- Cardiac Department, Aerospace Center Hospital, Beijing, China
- Cardiac Department, Peking University Aerospace School of Clinical Medicine, Beijing, China
| |
Collapse
|
6
|
Jiang J, Zhou M, Zhang Q, Shen H, Jia Y, Chen Y, Xu X, Jiang H. The Correlation Between Major Adverse Cardiovascular and Cerebrovascular Events (MACCE) and miR-142-3p in Maintenance Hemodialysis Patients With End-Stage Renal Disease. Semin Dial 2024; 37:373-379. [PMID: 39111739 DOI: 10.1111/sdi.13221] [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: 04/15/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Patients with end-stage renal disease (ESRD) on maintenance hemodialysis (MHD) are at high risk for major adverse cardiovascular and cerebrovascular events (MACCE), which are prone to be detrimental to patients' lives. Identifying risk factors for MACCE can help target measures to prevent or reduce the occurrence of MACCE. OBJECTIVE The aim was to investigate the correlation between miR-142-3p and MACCE in ESRD patients on MHD and to provide a new predictor for MACCE occurrence. METHODS Blood samples were collected from subjects to detect the expression of miR-142-3p using RT-qPCR. The correlation of miR-142-3p with HDL-C and hs-CRP was assessed by the Pearson method. The occurrence of MACCE in patients during the 36-month follow-up period was recorded. The clinical value of miR-142-3p in MACCE occurrence was analyzed by the Kaplan-Meier curve, multivariate logistic regression, and ROC curve. RESULTS In ESRD patients on MHD, miR-142-3p was downregulated, and it showed a positive correlation with HDL-C but a negative correlation with hs-CRP. The cumulative incidence of MACCE at 1, 2, and 3 years was 8.9%, 20.0%, and 30.4%, respectively. miR-142-3p levels were reduced in patients who developed MACCE and were associated with the cumulative incidence of MACCE. miR-142-3p was a risk factor for MACCE and showed a predictive value with specificity and sensitivity of 89.36% and 56.10%, respectively. CONCLUSIONS miR-142-3p was a risk factor of MACCE in ESRD patients undergoing MHD.
Collapse
Affiliation(s)
- Jiaxiang Jiang
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Meiling Zhou
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiong Zhang
- Nursing Department, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huajuan Shen
- Nursing Department, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanqing Jia
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanfang Chen
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiujun Xu
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hongfang Jiang
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Lee SY, Chao CT, Han DS, Chiang CK, Hung KY. A combined circulating microRNA panel predicts the risk of vascular calcification in community-dwelling older adults with age strata differences. Arch Gerontol Geriatr 2024; 120:105333. [PMID: 38262252 DOI: 10.1016/j.archger.2024.105333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 01/25/2024]
Abstract
BACKGROUND Older adults have a higher risk of developing vascular calcification (VC). Circulating miRNAs can be potential risk indicators. However, prior studies used single miRNA mostly, whereas miRNA panels were rarely evaluated. We aimed to examine whether a miRNA panel outperformed each miRNA alone, and analyzed whether advanced age affected VC risk predictive performance offered by the miRNA panel. METHODS We prospectively enrolled older adults (age ≥65 years) during their annual health checkup in 2017, and examined their VC severity followed by analyzing sera for VC regulatory miRNAs (miR-125b-5p, miR-125b-3p, and miR-378a-3p). We used multiple regression analyses to determine associations between each miRNA or a 3-combind panel and VC risk, followed by area under the receiver-operating-characteristics curve (AUROC) analysis. Participants were further divided to those of 65-75 and ≥75 years for comparison. RESULTS From 199 older adults screened, 169 (median age, 73.3 years) with available calcification assessment were analyzed, among whom 74.6 % having VC. Those with VC had significantly lower circulating miR-125b-5p, miR-125b-3p, and miR-378a-3p levels than those without. Regression analyses showed that the 3-combined miRNA panel exhibited significant associations with VC risk, with significantly higher AUROC than those of models based on individual miRNA. Importantly, in those ≥75 years, the miRNA-predicted risk of VC was more prominent than that in the 65-75 years group. CONCLUSION A miRNA panel for VC risk prediction might outperform individual miRNA alone in older adults, and advanced age modified the association between circulating miRNAs and the risk of VC.
Collapse
Affiliation(s)
- Szu-Ying Lee
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital Yunlin branch, Yunlin County, Taiwan; Division of Nephrology, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Ter Chao
- Division of Nephrology, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan City, Taiwan.
| | - Der-Sheng Han
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Integrative diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuan-Yu Hung
- Division of Nephrology, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| |
Collapse
|
8
|
Lu KC, Hung KC, Liao MT, Shih LJ, Chao CT. Vascular Calcification Heterogeneity from Bench to Bedside: Implications for Manifestations, Pathogenesis, and Treatment Considerations. Aging Dis 2024; 16:683-692. [PMID: 38739930 PMCID: PMC11964443 DOI: 10.14336/ad.2024.0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/20/2024] [Indexed: 05/16/2024] Open
Abstract
Vascular calcification (VC) is the ectopic deposition of calcium-containing apatite within vascular walls, exhibiting a high prevalence in older adults, and those with diabetes or chronic kidney disease. VC is a subclinical cardiovascular risk trait that increases mortality and functional deterioration. However, effective treatments for VC remain largely unavailable despite multiple attempts. Part of this therapeutic nihilism results from the failure to appreciate the diversity of VC as a pathological complex, with unforeseeable variations in morphology, risk associates, and anatomical and molecular pathogenesis, affecting clinical management strategies. VC should not be considered a homogeneous pathology because accumulating evidence refutes its conceptual and content uniformity. Here, we summarize the pathophysiological sources of VC heterogeneity from the intersecting pathways and networks of cellular, subcellular, and molecular crosstalk. Part of these pathological connections are synergistic or mutually antagonistic. We then introduce clinical implications related to the VC heterogeneity concept. Even within the same individual, a specific artery may exhibit the strongest tendency for calcification compared with other arteries. The prognostic value of VC may only be detectable with a detailed characterization of calcification morphology and features. VC heterogeneity is also evident, as VC risk factors vary between different arterial segments and layers. Therefore, diagnostic and screening strategies for VC may be improved based on VC heterogeneity, including the use of radiomics. Finally, pursuing a homogeneous treatment strategy is discouraged and we suggest a more rational approach by diversifying the treatment spectrum. This may greatly benefit subsequent efforts to identify effective VC therapeutics.
Collapse
Affiliation(s)
- Kuo-Cheng Lu
- Division of Nephrology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei, Taiwan.
| | - Kuo-Chin Hung
- Division of Nephrology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan.
- Department of Pharmacy, Tajen University, Pingtung, Taiwan.
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Hsinchu Branch, Hsinchu, Taiwan.
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Li-Jane Shih
- Department of Medical Laboratory, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan.
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan.
| | - Chia-Ter Chao
- Division of Nephrology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan.
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Division of Nephrology, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan.
- Center of Faculty Development, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
9
|
Jin J, Cheng M, Wu X, Zhang H, Zhang D, Liang X, Qian Y, Guo L, Zhang S, Bai Y, Xu J. Circulating miR-129-3p in combination with clinical factors predicts vascular calcification in hemodialysis patients. Clin Kidney J 2024; 17:sfae038. [PMID: 38524234 PMCID: PMC10960567 DOI: 10.1093/ckj/sfae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Indexed: 03/26/2024] Open
Abstract
Background Vascular calcification (VC) commonly occurs and seriously increases the risk of cardiovascular events and mortality in patients with hemodialysis. For optimizing individual management, we will develop a diagnostic multivariable prediction model for evaluating the probability of VC. Methods The study was conducted in four steps. First, identification of miRNAs regulating osteogenic differentiation of vascular smooth muscle cells (VSMCs) in calcified condition. Second, observing the role of miR-129-3p on VC in vitro and the association between circulating miR-129-3p and VC in hemodialysis patients. Third, collecting all indicators related to VC as candidate variables, screening predictors from the candidate variables by Lasso regression, developing the prediction model by logistic regression and showing it as a nomogram in training cohort. Last, verifying predictive performance of the model in validation cohort. Results In cell experiments, miR-129-3p was found to attenuate vascular calcification, and in human, serum miR-129-3p exhibited a negative correlation with vascular calcification, suggesting that miR-129-3p could be one of the candidate predictor variables. Regression analysis demonstrated that miR-129-3p, age, dialysis duration and smoking were valid factors to establish the prediction model and nomogram for VC. The area under receiver operating characteristic curve of the model was 0.8698. The calibration curve showed that predicted probability of the model was in good agreement with actual probability and decision curve analysis indicated better net benefit of the model. Furthermore, internal validation through bootstrap process and external validation by another independent cohort confirmed the stability of the model. Conclusion We build a diagnostic prediction model and present it as an intuitive tool based on miR-129-3p and clinical indicators to evaluate the probability of VC in hemodialysis patients, facilitating risk stratification and effective decision, which may be of great importance for reducing the risk of serious cardiovascular events.
Collapse
Affiliation(s)
- Jingjing Jin
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Meijuan Cheng
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Xueying Wu
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Haixia Zhang
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Dongxue Zhang
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Xiangnan Liang
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Yuetong Qian
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Liping Guo
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Shenglei Zhang
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Yaling Bai
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Jinsheng Xu
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| |
Collapse
|
10
|
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
|
11
|
Wu J, Wan M, Jiang Z, Gong W, Zhou X. lncRNA FAS-AS1 served as a diagnostic biomarker of end-stage renal disease and mediated vascular calcification via regulating oxidative stress and inflammation. Gene 2024; 896:148035. [PMID: 38013128 DOI: 10.1016/j.gene.2023.148035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/09/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE Vascular calcification is a frequently occurring complication of end-stage renal disease (ESRD). This study focused on the significance of long non-coding RNA Fas cell surface death receptor-antisense 1(lncRNA FAS-AS1) in ESRD-related vascular calcification aiming to explore a potential biomarker for the detection. METHODS The study enrolled 65 healthy individuals, 79 ESRD patients (48 patients with vascular calcification), and 93 early-stage (I-IV) chronic kidney disease (CKD) patients. The expression of FAS-AS1 in serum was evaluated by real-time quantitative polymerase chain reaction (PCR). The diagnostic potential of FAS-AS1 was assessed in discriminating ESRD patients, vascular calcification, and the severity of vascular calcification. In vitro, the vascular smooth muscle cells (VSMCs) were treated with a hyperphosphatemia medium to evaluate the effect of FAS-AS1 on VSMCs calcification. RESULTS Elevated serum FAS-AS1 was observed in ESRD patients, which could discriminate from healthy individuals and early-stage CKD patients. FAS-AS1 was associated with the development of ESRD and the occurrence of vascular calcification. FAS-AS1 was also upregulated in vascular calcification patients, especially the patients with severe calcification, which showed diagnostic significance in evaluating vascular calcification degrees. Calcified VSMCs showed significantly increased levels of Ca2+, reactive oxygen species (ROS), tumor necrosis factor-α (TNF-α), and interleukin 6 (IL-6), which was attenuated by silencing FAS-AS1. CONCLUSIONS FAS-AS1 discriminated ERSD patients and was associated with the occurrence of vascular calcification. The knockdown of FAS-AS1 suppressed hyperphosphatemia-induced vascular calcification via alleviating oxidative stress and inflammation.
Collapse
Affiliation(s)
- Jiaqi Wu
- Department of In-Patient Ultrasound, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Ming Wan
- Department of In-Patient Ultrasound, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Zhaopeng Jiang
- Department of In-Patient Ultrasound, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Wushuang Gong
- Department of In-Patient Ultrasound, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Xianli Zhou
- Department of In-Patient Ultrasound, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
12
|
Chao CT, Kuo FC, Lin SH. Epigenetically regulated inflammation in vascular senescence and renal progression of chronic kidney disease. Semin Cell Dev Biol 2024; 154:305-315. [PMID: 36241561 DOI: 10.1016/j.semcdb.2022.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/08/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
Abstract
Chronic kidney disease (CKD) and its complications, including vascular senescence and progressive renal fibrosis, are associated with inflammation. Vascular senescence, in particular, has emerged as an instrumental mediator of vascular inflammation that potentially worsens renal function. Epigenetically regulated inflammation involving histone modification, DNA methylation, actions of microRNAs and other non-coding RNAs, and their reciprocal reactions during vascular senescence and inflammaging are underappreciated. Their synergistic effects can contribute to CKD progression. Vascular senotherapeutics or pharmacological anti-senescent therapies based on epigenetic machineries can therefore be plausible options for ameliorating vascular aging and even halting the worsening of renal fibrosis. These include histone deacetylase modulators, histone methyltransferase modulators, other histone modification effectors, DNA methyltransferase inhibitors, telomerase reverse transcriptase enhancers, microRNA mimic delivery, and small molecules with microRNA-regulating potentials. Some of these molecules have already been tested and have shown anecdotal evidence for treating uremic vasculopathy and renal fibrosis, supporting the feasibility of this approach.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Nephrology division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Feng-Chih Kuo
- Division of Endocrinology, Department of Internal Medicine, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hua Lin
- Nephrology division, Department of Internal Medicine, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
13
|
Chao CT, Liao MT, Wu CK. Left Ventricular Hypertrophy Geometry and Vascular Calcification Co-Modify the Risk of Cardiovascular Mortality in Patients with End-Stage Kidney Disease: A Retrospective Cohort Study. J Atheroscler Thromb 2023; 30:1242-1254. [PMID: 36567124 PMCID: PMC10499460 DOI: 10.5551/jat.63870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/01/2022] [Indexed: 12/25/2022] Open
Abstract
AIM Patients with end-stage kidney disease (ESKD) have an unparalleled risk of left ventricular hypertrophy (LVH) and vascular calcification (VC), both of which introduce excessive cardiovascular risk. However, it remains unclear whether LVH geometry co-modulates cardiovascular outcomes with VC in this population. METHODS A retrospective cohort study was conducted. Patients with ESKD requiring chronic hemodialysis were identified from Shin Kong Wu Ho-Su Memorial Hospital between October and December 2018, with echocardiographic LVH geometry and aortic arch calcification (AoAC) determined. They were divided into four groups according to AoAC severity and eccentric or concentric LVH. We used Kaplan-Meier analysis and Cox proportional hazard regression to analyze their cardiovascular and all-cause mortality after multivariate adjustment. RESULTS Overall, 223 patients with ESKD with LVH were analyzed, among whom 29.1%, 23.3%, 25.1%, and 22.4% had non-to-mild AoAC with eccentric and concentric LVH and moderate-to-severe AoAC with eccentric and concentric LVH, respectively. After 3.5 years of follow-up, patients with ESKD with moderate-to-severe AoAC and concentric LVH had a significantly higher risk of cardiovascular mortality than those with non-to-mild AoAC and eccentric LVH (hazard ratio 3.35, p=0.002). However, those with moderate-to-severe AoAC but eccentric LVH did not have higher cardiovascular mortality. Similarly, patients with ESKD with moderate-to-severe AoAC and concentric LVH had a significantly higher all-cause mortality than those with non-to-mild AoAC and eccentric LVH, whereas the other two groups did not have higher risk. CONCLUSION LVH geometry could help stratify the risk of patients with ESKD when they had severe VC, and co-existing severe VC and concentric LVH aggravated cardiovascular risk.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Nephrology division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan
| | - Chung-Kuan Wu
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
14
|
Kuscu C, Mallisetty Y, Naik S, Han Z, Berta CJ, Kuscu C, Kovesdy CP, Sumida K. Circulating microRNA Profiles for Premature Cardiovascular Death in Patients with Kidney Failure with Replacement Therapy. J Clin Med 2023; 12:5010. [PMID: 37568412 PMCID: PMC10419472 DOI: 10.3390/jcm12155010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
INTRODUCTION Patients with kidney failure with replacement therapy (KFRT) suffer from a disproportionately high cardiovascular disease burden. Circulating small non-coding RNAs (c-sncRNAs) have emerged as novel epigenetic regulators and are suggested as novel biomarkers and therapeutic targets for cardiovascular disease; however, little is known about the associations of c-sncRNAs with premature cardiovascular death in KFRT. METHODS In a pilot case-control study of 50 hemodialysis patients who died of cardiovascular events as cases, and 50 matched hemodialysis controls who remained alive during a median follow-up of 2.0 years, we performed c-sncRNAs profiles using next-generation sequencing to identify differentially expressed circulating microRNAs (c-miRNAs) between the plasma of cases and that of controls. mRNA target prediction and pathway enrichment analysis were performed to examine the functional relevance of differentially expressed c-miRNAs to cardiovascular pathophysiology. The association of differentially expressed c-miRNAs with cardiovascular mortality was examined using multivariable conditional logistic regression. RESULTS The patient characteristics were similar between cases and controls, with a mean age of 63 years, 48% male, and 54% African American in both groups. We detected a total of 613 miRNAs in the plasma, among which five miRNAs (i.e., miR-129-1-5p, miR-500b-3p, miR-125b-1-3p, miR-3648-2-5p, and miR-3150b-3p) were identified to be differentially expressed between cases and controls with cut-offs of p < 0.05 and log2 fold-change (log2FC) > 1. When using more stringent cut-offs of p-adjusted < 0.05 and log2FC > 1, only miR-129-1-5p remained significantly differentially expressed, with higher levels of miR-129-1-5p in the cases than in the controls. The pathway enrichment analysis using predicted miR-129-1-5p mRNA targets demonstrated enrichment in adrenergic signaling in cardiomyocytes, arrhythmogenic right ventricular cardiomyopathy, and oxytocin signaling pathways. In parallel, the circulating miR-129-1-5p levels were significantly associated with the risk of cardiovascular death (adjusted OR [95% CI], 1.68 [1.01-2.81] for one increase in log-transformed miR-129-1-5p counts), independent of potential confounders. CONCLUSIONS Circulating miR-129-1-5p may serve as a novel biomarker for premature cardiovascular death in KFRT.
Collapse
Affiliation(s)
- Canan Kuscu
- Transplant Research Institute, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.N.); (C.K.)
| | - Yamini Mallisetty
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.M.); (Z.H.); (C.J.B.); (C.P.K.)
| | - Surabhi Naik
- Transplant Research Institute, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.N.); (C.K.)
| | - Zhongji Han
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.M.); (Z.H.); (C.J.B.); (C.P.K.)
| | - Caleb J. Berta
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.M.); (Z.H.); (C.J.B.); (C.P.K.)
| | - Cem Kuscu
- Transplant Research Institute, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (S.N.); (C.K.)
| | - Csaba P. Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.M.); (Z.H.); (C.J.B.); (C.P.K.)
- Nephrology Section, Memphis VA Medical Center, Memphis, TN 38104, USA
| | - Keiichi Sumida
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (Y.M.); (Z.H.); (C.J.B.); (C.P.K.)
| |
Collapse
|
15
|
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: 13] [Impact Index Per Article: 6.5] [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
|
16
|
Zang D, Li J, Zhou C. Clinical expression of microRNA-144-5p and its regulatory effect on renal function in uremia. Ther Apher Dial 2023; 27:246-252. [PMID: 35997718 DOI: 10.1111/1744-9987.13922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The study commits to probing the clinical expression of microRNA-144-5p (miR-144-5p) and its modulatory effect on the renal function of uremia. METHODS Levels of blood urea nitrogen (BUN), β2-microglobulin (β2-MG), serum creatinine (Scr), blood calcium (Ca), phosphorus (P), and intact parathyroid hormone (iPTH) and miR-144-5p expression in serum of uremia patients were detected. The correlation among miR-144-5p expression with BUN, β2-MG, Scr, Ca, P, and iPTH levels in uremic patients was analyzed. The rats were injected with miR-144-5p agomir to detect the change of BUN, Scr, β2-MG, Scr, Ca, P, and iPTH levels in uremic rats. RESULTS miR-144-5p expression in uremic patients was negatively correlated with BUN, Scr, β2-MG, P, and iPTH levels, and positively correlated with free Ca concentration in blood. miR-144-5p elevation reduced BUN, Scr, β2-MG, P, and iPTH levels, and increased free Ca concentration in blood in uremic rats. CONCLUSION miR-144-5p is lowly expressed, and miR-144-5p has a regulatory effect on renal function in uremia.
Collapse
Affiliation(s)
- Dong Zang
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western, Beijing, China
| | - Junyi Li
- Department of Clinical Laboratory, Beijing Maternal and Child Health Care Hospital Yanqing District, Beijing, China
| | - Chuanyan Zhou
- Department of Clinical Laboratory, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Chao CT, Hung KY. Vascular frailty, a proposal for new frailty type: A narrative review. Kaohsiung J Med Sci 2023; 39:318-325. [PMID: 36866657 DOI: 10.1002/kjm2.12664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 03/04/2023] Open
Abstract
Frailty is the incremental accumulation of minute defects that progressively impair health and performance. Frailty is commonly observed in older adults; however, secondary frailty may also occur in patients with metabolic disorders or major organ failure. In addition to physical frailty, several distinct types of frailty have been identified, including oral, cognitive, and social frailty, each of which is of practical importance. This nomenclature suggests that detailed descriptions of frailty can potentially advance relevant researches. In this narrative review, we first summarize the clinical value and plausible biological origin of frailty, as well as how to appropriately assess it using physical frailty phenotypes and frailty indexes. In the second part, we discuss the issue of vascular tissue as a relatively underappreciated organ whose pathologies contribute to the development of physical frailty. Moreover, when vascular tissue undergoes degeneration, it exhibits vulnerability to subtle injuries and manifests a unique phenotype amenable to clinical assessment prior to or accompanying physical frailty development. Finally, we propose that vascular frailty, based on an extensive set of experimental and clinical evidence, can be considered a new frailty type that requires our attention. We also outline potential methods for the operationalization of vascular frailty. Further studies are required to validate our claim and sharpen the spectrum of this degenerative phenotype.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan.,Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuan-Yu Hung
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
18
|
The context-dependent role of transforming growth factor-β/miR-378a-3p/connective tissue growth factor in vascular calcification: a translational study. Aging (Albany NY) 2023; 15:830-845. [PMID: 36787443 PMCID: PMC9970315 DOI: 10.18632/aging.204518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Vascular calcification (VC) constitutes an important vascular pathology with prognostic importance. The pathogenic role of transforming growth factor-β (TGF-β) in VC remains unclear, with heterogeneous findings that we aimed to evaluate using experimental models and clinical specimens. METHODS Two approaches, exogenous administration and endogenous expression upon osteogenic media (OM) exposure, were adopted. Aortic smooth muscle cells (ASMCs) were subjected to TGF-β1 alone, OM alone, or both, with calcification severity determined. We evaluated miR-378a-3p and TGF-β1 effectors (connective tissue growth factor; CTGF) at different periods of calcification. Results were validated in an ex vivo model and further in sera from older adults without or with severe aortic arch calcification. RESULTS TGF-β1 treatment induced a significant dose-responsive increase in ASMC calcification without or with OM at the mature but not early or mid-term VC period. On the other hand, OM alone induced VC accompanied by suppressed TGF-β1 expressions over time; this phenomenon paralleled the declining miR-378a-3p and CTGF expressions since early VC. TGF-β1 treatment led to an upregulation of CTGF since early VC but not miR-378a-3p until mid-term VC, while miR-378a-3p overexpression suppressed CTGF expressions without altering TGF-β1 levels. The OM-induced down-regulation of TGF-β1 and CTGF was also observed in the ex vivo models, with compatible results identified from human sera. CONCLUSIONS We showed that TGF-β1 played a context-dependent role in VC, involving a time-dependent self-regulatory loop of TGF-β1/miR-378a-3p/CTGF signaling. Our findings may assist subsequent studies in devising potential therapeutics against VC.
Collapse
|
19
|
Sutton NR, Malhotra R, Hilaire C, Aikawa E, Blumenthal RS, Gackenbach G, Goyal P, Johnson A, Nigwekar SU, Shanahan CM, Towler DA, Wolford BN, Chen Y. Molecular Mechanisms of Vascular Health: Insights From Vascular Aging and Calcification. Arterioscler Thromb Vasc Biol 2023; 43:15-29. [PMID: 36412195 PMCID: PMC9793888 DOI: 10.1161/atvbaha.122.317332] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Abstract
Cardiovascular disease is the most common cause of death worldwide, especially beyond the age of 65 years, with the vast majority of morbidity and mortality due to myocardial infarction and stroke. Vascular pathology stems from a combination of genetic risk, environmental factors, and the biologic changes associated with aging. The pathogenesis underlying the development of vascular aging, and vascular calcification with aging, in particular, is still not fully understood. Accumulating data suggests that genetic risk, likely compounded by epigenetic modifications, environmental factors, including diabetes and chronic kidney disease, and the plasticity of vascular smooth muscle cells to acquire an osteogenic phenotype are major determinants of age-associated vascular calcification. Understanding the molecular mechanisms underlying genetic and modifiable risk factors in regulating age-associated vascular pathology may inspire strategies to promote healthy vascular aging. This article summarizes current knowledge of concepts and mechanisms of age-associated vascular disease, with an emphasis on vascular calcification.
Collapse
Affiliation(s)
- Nadia R. Sutton
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Rajeev Malhotra
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Cynthia Hilaire
- Division of Cardiology, Departments of Medicine and Bioengineering, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, 1744 BSTWR, 200 Lothrop St, Pittsburgh, PA, 15260 USA
| | - Elena Aikawa
- Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Roger S. Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease; Baltimore, MD
| | - Grace Gackenbach
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Parag Goyal
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Adam Johnson
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Sagar U. Nigwekar
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Catherine M. Shanahan
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| | - Dwight A. Towler
- Department of Medicine | Endocrine Division and Pak Center for Mineral Metabolism Research, UT Southwestern Medical Center, Dallas, TX USA
| | - Brooke N. Wolford
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham and Research Department, Veterans Affairs Birmingham Medical Center, Birmingham, AL, USA
| |
Collapse
|
20
|
Ren SC, Mao N, Yi S, Ma X, Zou JQ, Tang X, Fan JM. Vascular Calcification in Chronic Kidney Disease: An Update and Perspective. Aging Dis 2022; 13:673-697. [PMID: 35656113 PMCID: PMC9116919 DOI: 10.14336/ad.2021.1024] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease is a devastating condition resulting from irreversible loss of nephron numbers and function and leading to end-stage renal disease and mineral disorders. Vascular calcification, an ectopic deposition of calcium-phosphate salts in blood vessel walls and heart valves, is an independent risk factor of cardiovascular morbidity and mortality in chronic kidney disease. Moreover, aging and related metabolic disorders are essential risk factors for chronic kidney disease and vascular calcification. Marked progress has been recently made in understanding and treating vascular calcification in chronic kidney disease. However, there is a paucity of systematic reviews summarizing this progress, and investigating unresolved issues is warranted. In this systematic review, we aimed to overview the underlying mechanisms of vascular calcification in chronic kidney diseases and discuss the impact of chronic kidney disease on the pathophysiology of vascular calcification. Additionally, we summarized potential clinical diagnostic biomarkers and therapeutic applications for vascular calcification with chronic kidney disease. This review may offer new insights into the pathogenesis, diagnosis, and therapeutic intervention of vascular calcification.
Collapse
Affiliation(s)
- Si-Chong Ren
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
- Center for Translational Medicine, Sichuan Academy of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Mao
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Si Yi
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| | - Xin Ma
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Jia-Qiong Zou
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jun-Ming Fan
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| |
Collapse
|
21
|
Deep Learning-Assisted Repurposing of Plant Compounds for Treating Vascular Calcification: An In Silico Study with Experimental Validation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4378413. [PMID: 35035662 PMCID: PMC8754599 DOI: 10.1155/2022/4378413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/24/2021] [Accepted: 11/13/2021] [Indexed: 12/13/2022]
Abstract
Background Vascular calcification (VC) constitutes subclinical vascular burden and increases cardiovascular mortality. Effective therapeutics for VC remains to be procured. We aimed to use a deep learning-based strategy to screen and uncover plant compounds that potentially can be repurposed for managing VC. Methods We integrated drugome, interactome, and diseasome information from Comparative Toxicogenomic Database (CTD), DrugBank, PubChem, Gene Ontology (GO), and BioGrid to analyze drug-disease associations. A deep representation learning was done using a high-level description of the local network architecture and features of the entities, followed by learning the global embeddings of nodes derived from a heterogeneous network using the graph neural network architecture and a random forest classifier established for prediction. Predicted results were tested in an in vitro VC model for validity based on the probability scores. Results We collected 6,790 compounds with available Simplified Molecular-Input Line-Entry System (SMILES) data, 11,958 GO terms, 7,238 diseases, and 25,482 proteins, followed by local embedding vectors using an end-to-end transformer network and a node2vec algorithm and global embedding vectors learned from heterogeneous network via the graph neural network. Our algorithm conferred a good distinction between potential compounds, presenting as higher prediction scores for the compound categories with a higher potential but lower scores for other categories. Probability score-dependent selection revealed that antioxidants such as sulforaphane and daidzein were potentially effective compounds against VC, while catechin had low probability. All three compounds were validated in vitro. Conclusions Our findings exemplify the utility of deep learning in identifying promising VC-treating plant compounds. Our model can be a quick and comprehensive computational screening tool to assist in the early drug discovery process.
Collapse
|
22
|
Wu PY, Lee SY, Chang KV, Chao CT, Huang JW. Gender-Related Differences in Chronic Kidney Disease-Associated Vascular Calcification Risk and Potential Risk Mediators: A Scoping Review. Healthcare (Basel) 2021; 9:979. [PMID: 34442116 PMCID: PMC8394860 DOI: 10.3390/healthcare9080979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Vascular calcification (VC) involves the deposition of calcium apatite in vascular intima or media. Individuals of advanced age, having diabetes mellitus or chronic kidney disease (CKD) are particularly at risk. The pathogenesis of CKD-associated VC evolves considerably. The core driver is the phenotypic change involving vascular wall constituent cells toward manifestations similar to that undergone by osteoblasts. Gender-related differences are observed regarding the expressions of osteogenesis-regulating effectors, and presumably the prevalence/risk of CKD-associated VC exhibits gender-related differences as well. Despite the wealth of data focusing on gender-related differences in the risk of atherosclerosis, few report whether gender modifies the risk of VC, especially CKD-associated cases. We systematically identified studies of CKD-associated VC or its regulators/modifiers reporting data about gender distributions, and extracted results from 167 articles. A significantly higher risk of CKD-associated VC was observed in males among the majority of original investigations. However, substantial heterogeneity exists, since multiple large-scale studies yielded neutral findings. Differences in gender-related VC risk may result from variations in VC assessment methods, the anatomical segments of interest, study sample size, and even the ethnic origins of participants. From a biological perspective, plausible mediators of gender-related VC differences include body composition discrepancies, alterations involving lipid profiles, inflammatory severity, diversities in matrix Gla protein (MGP), soluble Klotho, vitamin D, sclerostin, parathyroid hormone (PTH), fibroblast growth factor-23 (FGF-23), and osteoprotegerin levels. Based on our findings, it may be inappropriate to monotonously assume that male patients with CKD are at risk of VC compared to females, and we should consider more background in context before result interpretation.
Collapse
Affiliation(s)
- Patrick Yihong Wu
- School of Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| | - Szu-Ying Lee
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin County 640, Taiwan; (S.-Y.L.); (J.-W.H.)
| | - Ke-Vin Chang
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan;
| | - Chia-Ter Chao
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan
| | - Jenq-Wen Huang
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin County 640, Taiwan; (S.-Y.L.); (J.-W.H.)
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| |
Collapse
|
23
|
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: 27] [Impact Index Per Article: 6.8] [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
|
24
|
Chao CT, Lin SH. Uremic Toxins and Frailty in Patients with Chronic Kidney Disease: A Molecular Insight. Int J Mol Sci 2021; 22:ijms22126270. [PMID: 34200937 PMCID: PMC8230495 DOI: 10.3390/ijms22126270] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
The accumulation of uremic toxins (UTs) is a prototypical manifestation of uremic milieu that follows renal function decline (chronic kidney disease, CKD). Frailty as a potential outcome-relevant indicator is also prevalent in CKD. The intertwined relationship between uremic toxins, including small/large solutes (phosphate, asymmetric dimethylarginine) and protein-bound ones like indoxyl sulfate (IS) and p-cresyl sulfate (pCS), and frailty pathogenesis has been documented recently. Uremic toxins were shown in vitro and in vivo to induce noxious effects on many organ systems and likely influenced frailty development through their effects on multiple preceding events and companions of frailty, such as sarcopenia/muscle wasting, cognitive impairment/cognitive frailty, osteoporosis/osteodystrophy, vascular calcification, and cardiopulmonary deconditioning. These organ-specific effects may be mediated through different molecular mechanisms or signal pathways such as peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), mitogen-activated protein kinase (MAPK) signaling, aryl hydrocarbon receptor (AhR)/nuclear factor-κB (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), Runt-related transcription factor 2 (RUNX2), bone morphogenic protein 2 (BMP2), osterix, Notch signaling, autophagy effectors, microRNAs, and reactive oxygen species induction. Anecdotal clinical studies also suggest that frailty may further accelerate renal function decline, thereby augmenting the accumulation of UTs in affected individuals. Judging from these threads of evidence, management strategies aiming for uremic toxin reduction may be a promising approach for frailty amelioration in patients with CKD. Uremic toxin lowering strategies may bear the potential of improving patients’ outcomes and restoring their quality of life, through frailty attenuation. Pathogenic molecule-targeted therapeutics potentially disconnect the association between uremic toxins and frailty, additionally serving as an outcome-modifying approach in the future.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan;
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100255, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Shih-Hua Lin
- Nephrology Division, Department of Internal Medicine, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: or
| |
Collapse
|
25
|
Düsing P, Zietzer A, Goody PR, Hosen MR, Kurts C, Nickenig G, Jansen F. Vascular pathologies in chronic kidney disease: pathophysiological mechanisms and novel therapeutic approaches. J Mol Med (Berl) 2021; 99:335-348. [PMID: 33481059 PMCID: PMC7900031 DOI: 10.1007/s00109-021-02037-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/14/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of death in patients with chronic kidney disease (CKD). Both conditions are rising in incidence as well as prevalence, creating poor outcomes for patients and high healthcare costs. Recent data suggests CKD to be an independent risk factor for CVD. Accumulation of uremic toxins, chronic inflammation, and oxidative stress have been identified to act as CKD-specific alterations that increase cardiovascular risk. The association between CKD and cardiovascular mortality is markedly influenced through vascular alterations, in particular atherosclerosis and vascular calcification (VC). While numerous risk factors promote atherosclerosis by inducing endothelial dysfunction and its progress to vascular structural damage, CKD affects the medial layer of blood vessels primarily through VC. Ongoing research has identified VC to be a multifactorial, cell-mediated process in which numerous abnormalities like mineral dysregulation and especially hyperphosphatemia induce a phenotype switch of vascular smooth muscle cells to osteoblast-like cells. A combination of pro-calcifying stimuli and an impairment of inhibiting mechanisms like fetuin A and vitamin K-dependent proteins like matrix Gla protein and Gla-rich protein leads to mineralization of the extracellular matrix. In view of recent studies, intercellular communication pathways via extracellular vesicles and microRNAs represent key mechanisms in VC and thereby a promising field to a deeper understanding of the involved pathomechanisms. In this review, we provide an overview about pathophysiological mechanisms connecting CKD and CVD. Special emphasis is laid on vascular alterations and more recently discovered molecular pathways which present possible new therapeutic targets.
Collapse
Affiliation(s)
- Philip Düsing
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Andreas Zietzer
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Philip Roger Goody
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Mohammed Rabiul Hosen
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, Bonn, 53127, Germany
| | - Georg Nickenig
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Felix Jansen
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
26
|
Chao CT, Han DS, Huang JW. Circulating microRNA-125b Levels Are Associated With the Risk of Vascular Calcification in Healthy Community-Dwelling Older Adults. Front Cardiovasc Med 2021; 8:624313. [PMID: 33693036 PMCID: PMC7937626 DOI: 10.3389/fcvm.2021.624313] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/28/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Vascular calcification (VC) is a subclinical manifestation of vascular disease burden among older adults, conferring an elevated mortality risk. Biomarkers capable of detecting and risk-stratifying VC associated with advanced age remains unavailable, impeding our effort to provide optimal care to geriatric patients. Objectives: In this study, we aimed to investigate whether circulating miR-125b served as a potential indicator for VC in relatively healthy older adults. Methods: Community-dwelling older adults (age ≥65) were prospectively recruited during 2017, followed by clinical features documentation and VC rating based on aortic arch calcification (AAC) and abdominal aortic calcification (AbAC). Multiple logistic regression was done to evaluate the relationship between circulating miR-125b levels, VC presence and severity, followed by selecting the optimal cutoff point for VC diagnosis. Results: A total of 343 relatively healthy older adults (median age, 73.8 years; 40% male; 59.8% having AAC) were enrolled, with a median circulating miR-125b level of 0.012 (interquartile range, 0.003–0.037). Those with more severe AAC had progressively decreasing miR-125b levels (p<0.001). Multiple regression analyses showed that having higher miR-125b levels based on the median value were associated with a substantially lower risk of AAC [odds ratio (OR) 0.022, 95% confidence interval (CI) 0.011–0.044] compared to those having lower ones. An optimal cutoff of miR-125b for identifying AAC in older adults was 0.008, with a sensitivity and specificity of 0.86 and 0.80, respectively. Similar findings were obtained when using AbAC as the endpoint. Conclusions: We found that miR-125b serves as an independent indicator for VC in relatively healthy older adults, and may potentially be linked with VC pathophysiology.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan.,Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan.,Geriatric and Community Medicine Research Center, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan
| | - Der-Sheng Han
- Geriatric and Community Medicine Research Center, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan.,Department of Rehabilitation and Physical Medicine, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan
| | - Jenq-Wen Huang
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| |
Collapse
|
27
|
Tsai YT, Yeh HY, Chao CT, Chiang CK. Superoxide Dismutase 2 (SOD2) in Vascular Calcification: A Focus on Vascular Smooth Muscle Cells, Calcification Pathogenesis, and Therapeutic Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6675548. [PMID: 33728027 PMCID: PMC7935587 DOI: 10.1155/2021/6675548] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/27/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
Vascular calcification (VC) describes the pathophysiological phenotype of calcium apatite deposition within the vascular wall, leading to vascular stiffening and the loss of compliance. VC is never benign; the presence and severity of VC correlate closely with the risk of myocardial events and cardiovascular mortality in multiple at-risk populations such as patients with diabetes and chronic kidney disease. Mitochondrial dysfunction involving each of vascular wall constituents (endothelia and vascular smooth muscle cells (VSMCs)) aggravates various vascular pathologies, including atherosclerosis and VC. However, few studies address the pathogenic role of mitochondrial dysfunction during the course of VC, and mitochondrial reactive oxygen species (ROS) seem to lie in the pathophysiologic epicenter. Superoxide dismutase 2 (SOD2), through its preferential localization to the mitochondria, stands at the forefront against mitochondrial ROS in VSMCs and thus potentially modifies the probability of VC initiation or progression. In this review, we will provide a literature-based summary regarding the relationship between SOD2 and VC in the context of VSMCs. Apart from the conventional wisdom of attenuating mitochondrial ROS, SOD2 has been found to affect mitophagy and the formation of the autophagosome, suppress JAK/STAT as well as PI3K/Akt signaling, and retard vascular senescence, all of which underlie the beneficial influences on VC exerted by SOD2. More importantly, we outline the therapeutic potential of a novel SOD2-targeted strategy for the treatment of VC, including an ever-expanding list of pharmaceuticals and natural compounds. It is expected that VSMC SOD2 will become an important druggable target for treating VC in the future.
Collapse
Affiliation(s)
- You-Tien Tsai
- 1Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan
| | - Hsiang-Yuan Yeh
- 2School of Big Data Management, Soochow University, Taipei, Taiwan
| | - Chia-Ter Chao
- 1Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan
- 3Nephrology Division, Department of Internal Medicine, National Taiwan University School of Medicine, Taipei, Taiwan
- 4Graduate Institute of Toxicology, National Taiwan University School of Medicine, Taipei, Taiwan
| | - Chih-Kang Chiang
- 4Graduate Institute of Toxicology, National Taiwan University School of Medicine, Taipei, Taiwan
| |
Collapse
|
28
|
Chao CT, Lin SH. Uremic Vascular Calcification: The Pathogenic Roles and Gastrointestinal Decontamination of Uremic Toxins. Toxins (Basel) 2020; 12:toxins12120812. [PMID: 33371477 PMCID: PMC7767516 DOI: 10.3390/toxins12120812] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 12/16/2022] Open
Abstract
Uremic vascular calcification (VC) commonly occurs during advanced chronic kidney disease (CKD) and significantly increases cardiovascular morbidity and mortality. Uremic toxins are integral within VC pathogenesis, as they exhibit adverse vascular influences ranging from atherosclerosis, vascular inflammation, to VC. Experimental removal of these toxins, including small molecular (phosphate, trimethylamine-N-oxide), large molecular (fibroblast growth factor-23, cytokines), and protein-bound ones (indoxyl sulfate, p-cresyl sulfate), ameliorates VC. As most uremic toxins share a gut origin, interventions through gastrointestinal tract are expected to demonstrate particular efficacy. The “gastrointestinal decontamination” through the removal of toxin in situ or impediment of toxin absorption within the gastrointestinal tract is a practical and potential strategy to reduce uremic toxins. First and foremost, the modulation of gut microbiota through optimizing dietary composition, the use of prebiotics or probiotics, can be implemented. Other promising strategies such as reducing calcium load, minimizing intestinal phosphate absorption through the optimization of phosphate binders and the inhibition of gut luminal phosphate transporters, the administration of magnesium, and the use of oral toxin adsorbent for protein-bound uremic toxins may potentially counteract uremic VC. Novel agents such as tenapanor have been actively tested in clinical trials for their potential vascular benefits. Further advanced studies are still warranted to validate the beneficial effects of gastrointestinal decontamination in the retardation and treatment of uremic VC.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan;
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Shih-Hua Lin
- Department of Internal Medicine, Tri-Service General Hospital and National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence:
| |
Collapse
|
29
|
Ciceri P, Tettamanti G, Galassi A, Magagnoli L, Fabresse N, Alvarez JC, Massy ZA, Messa P, Cozzolino M. Pro-calcifying analysis of uraemic serum from patients treated with medium cut-off membrane in a prospective, cross-over study. Clin Kidney J 2020; 14:1798-1807. [PMID: 34221387 PMCID: PMC8243281 DOI: 10.1093/ckj/sfaa216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022] Open
Abstract
Background The retention of a large number of solutes that are normally excreted or metabolized by the kidney is responsible for the symptoms typical in uraemic patients. These molecules are defined as uraemic toxins and can be classified into three groups: small water-soluble molecules, middle molecules and protein-bound toxins. Recently, efforts were put towards developing dialysis membranes that allow the removal of large middle molecules without clinically relevant albumin loss. These membranes are the medium cut-off (MCO) membranes that allow the removal of middle molecules up to ∼50 000 Da. Methods We performed a prospective, open-label, controlled, cross-over pilot study comparing expanded haemodialysis (HDx) (novel MCO membrane Theranova 400) and conventional haemodialysis (HD) in 20 prevalent HD patients. Ten patients used conventional HD high-flux dialyser and 10 patients used HDx for 3 months; later the patients switched and received the other treatment for a further 3 months. We then analysed the pro-calcifying effect of uraemic serum in a model of high phosphate(Pi)-induced calcification in vascular smooth muscle cells (VSMCs). Results In this study, every patient was the control of himself and, interestingly, we found a tendency of less pro-calcifying potential from HDx-treated patients' serum compared with HD. Studying pathogenetic processes involved in high Pi-induced calcium deposition, we found that uraemic serum of HDx-treated patients induced less VSMC necrosis compared with uraemic serum of HD patients. Nevertheless, no differences were found between the different dialytic treatments in the serum potential to induce apoptosis and to modulate the expression of a panel of genes involved in VSMC simil-osteoblastic differentiation such as bone morphogenetic protein 2, runt-related transcription factor 2, osteocalcin, matrix Gla protein, osteopontin, elastin and collagen I α1. In an effort to characterize the difference in uraemic toxin profile during the two different dialytic treatments, we measured a panel of 10 uraemic toxins and 3 precursors, finding a significant increased removal during HDx of 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid, tryptophane and some of its metabolites, such as 3-indoxyl sulphate, indole 3-acetic acid and kynurenine. Conclusions These preliminary data are promising, although larger patients' groups are needed to better understand the effects of HDx on vascular calcification.
Collapse
Affiliation(s)
- Paola Ciceri
- Department of Nephrology, Dialysis and Renal Transplant, Renal Research Laboratory, Fondazione Ca' Granda IRCCS, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgia Tettamanti
- Department of Health Sciences, Renal Division, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Andrea Galassi
- Department of Health Sciences, Renal Division, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Lorenza Magagnoli
- Department of Health Sciences, Renal Division, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Nicolas Fabresse
- Laboratory of Pharmacology and Toxicology, CHU Raymond Poincare, Garches, France
| | - Jean-Claude Alvarez
- Laboratory of Pharmacology and Toxicology, CHU Raymond Poincare, Garches, France.,INSERM U-1173, UFR des Sciences de la Santé Simone Veil, Université Paris-Saclay (Versailles-Saint-Quentin-en-Yvelines), Montigny le Bretonneux, France
| | - Ziad A Massy
- Division of Nephrology, Ambroise Paré University Hospital, APHP, Boulogne-Billancourt/Paris, Paris, France.,Centre for Research in Epidemiology and Population Health (CESP), INSERM UMRS 1018, Université Paris-Saclay, Université Versailles Saint Quentin (UVSQ), Villejuif, France
| | - Piergiorgio Messa
- Department of Nephrology, Dialysis and Renal Transplant, Renal Research Laboratory, Fondazione Ca' Granda IRCCS, Ospedale Maggiore Policlinico, Milan, Italy
| | - Mario Cozzolino
- Department of Health Sciences, Renal Division, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| |
Collapse
|
30
|
Chao CT, Yeh HY, Tsai YT, Yuan TH, Liao MT, Huang JW, Chen HW. Astaxanthin Counteracts Vascular Calcification In Vitro Through an Early Up-Regulation of SOD2 Based on a Transcriptomic Approach. Int J Mol Sci 2020; 21:ijms21228530. [PMID: 33198315 PMCID: PMC7698184 DOI: 10.3390/ijms21228530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification (VC) is a critical contributor to the rising cardiovascular risk among at-risk populations such as those with diabetes or renal failure. The pathogenesis of VC involves an uprising of oxidative stress, for which antioxidants can be theoretically effective. However, astaxanthin, a potent antioxidant, has not been tested before for the purpose of managing VC. To answer this question, we tested the efficacy of astaxanthin against VC using the high phosphate (HP)-induced vascular smooth muscle cell (VSMC) calcification model. RNAs from treated groups underwent Affymetrix microarray screening, with intra-group consistency and inter-group differential expressions identified. Candidate hub genes were selected, followed by validation in experimental models and functional characterization. We showed that HP induced progressive calcification among treated VSMCs, while astaxanthin dose-responsively and time-dependently ameliorated calcification severities. Transcriptomic profiling revealed that 3491 genes exhibited significant early changes during VC progression, among which 26 potential hub genes were selected based on closeness ranking and biologic plausibility. SOD2 was validated in the VSMC model, shown to drive the deactivation of cellular senescence and enhance antioxidative defenses. Astaxanthin did not alter intracellular reactive oxygen species (ROS) levels without HP, but significantly lowered ROS production in HP-treated VSMCs. SOD2 knockdown prominently abolished the anti-calcification effect of astaxanthin on HP-treated VSMCs, lending support to our findings. In conclusion, we demonstrated for the first time that astaxanthin could be a potential candidate treatment for VC, through inducing the up-regulation of SOD2 early during calcification progression and potentially suppressing vascular senescence.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan; (C.-T.C.); (Y.-T.T.)
- Geriatric and Community Medicine Research Center, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| | - Hsiang-Yuan Yeh
- School of Big Data Management, Soochow University, Taipei 11102, Taiwan;
| | - You-Tien Tsai
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan; (C.-T.C.); (Y.-T.T.)
| | - Tzu-Hang Yuan
- Genome and Systems Biology Degree Program, Academia Sinica, Taipei 11529, Taiwan;
| | - Min-Tser Liao
- Department of Pediatrics, Armed Force Taoyuan General Hospital, Taoyuan County 32551, Taiwan;
| | - Jenq-Wen Huang
- Nephrology division, Department of Internal Medicine, National Taiwan University Hospital YunLin Branch, YunLin County 640203, Taiwan
- Correspondence: ; Tel.: +886-5-5323911 (ext. 5675)
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| |
Collapse
|
31
|
Lee S, Chao C, Huang J, Huang K. Vascular Calcification as an Underrecognized Risk Factor for Frailty in 1783 Community-Dwelling Elderly Individuals. J Am Heart Assoc 2020; 9:e017308. [PMID: 32875940 PMCID: PMC7727009 DOI: 10.1161/jaha.120.017308] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022]
Abstract
Background Vascular calcification (VC) is associated with high morbidity and mortality among older adults, a population that exhibits a higher tendency for developing frailty at the same time. Whether VC serves as a risk factor for the development of frailty in this population remains unclear. Methods and Results We analyzed a prospectively assembled cohort of community-dwelling older adults between 2014 and 2017 (n=1783). Frailty and prefrailty were determined on the basis of the Study of Osteoporotic Fractures criteria, and VC was measured using semiquantitative aortic arch calcification (AAC) and abdominal aortic calcification scoring. We conducted multiple logistic regression with prefrailty or frailty as the dependent variable, incorporating sociodemographic profiles, comorbidities, medications, laboratory data, AAC status/severity, and other geriatric phenotypes. Among all participants, 327 (18.3%) exhibited either prefrailty (15.3%) or frailty (3.1%), and 648 (36.3%) exhibited AAC. After adjusting for multiple confounders, we found that AAC incidence was associated with a substantially higher probability of prefrailty or frailty (odds ratio [OR], 11.9; 95% CI, 7.9-15.4), with a dose-responsive relationship (OR for older adults with AAC categories 1, 2, and 3 was 9.3, 13.6, and 52.5, respectively). Similar association was observed for older adults with abdominal aortic calcification (OR, 5.0; 95% CI, 1.3-19.5), and might be replicable in another cohort of patients with end-stage renal disease. Conclusions Severity of VC exhibited a linear positive relationship with frailty in older adults. Our findings suggest that a prompt diagnosis and potential management of VC may assist in risk mitigation for patients with frailty.
Collapse
Affiliation(s)
- Szu‐Ying Lee
- Nephrology Division, Department of Internal MedicineNational Taiwan University Hospital Yunlin BranchYunlin CountyTaiwan
| | - Chia‐Ter Chao
- Nephrology Division, Department of Internal MedicineNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
- Geriatric and Community Medicine Research CenterNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
- Graduate Institute of ToxicologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Jenq‐Wen Huang
- Nephrology Division, Department of Internal MedicineNational Taiwan University Hospital Yunlin BranchYunlin CountyTaiwan
| | - Kuo‐Chin Huang
- Nephrology Division, Department of Internal MedicineNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
- Geriatric and Community Medicine Research CenterNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
| |
Collapse
|
32
|
Wu BW, Guo JD, Wu MS, Liu Y, Lu M, Zhou YH, Han HW. Osteoblast-derived lipocalin-2 regulated by miRNA-96-5p/Foxo1 advances the progression of Alzheimer's disease. Epigenomics 2020; 12:1501-1513. [PMID: 32901506 DOI: 10.2217/epi-2019-0215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: Alzheimer's disease (AD) is the most frequent cause of dementia and characterized by the accumulation of β-amyloid peptides in plaques and vessel walls. This study proposed a hypothesis of an inhibitory role of miR-96-5p in AD via regulating Foxo1. Methods & methods: AD mouse models were established by injecting with 1% pentobarbital. Results: Knockdown of miR-96-5p in the presence of naringin was shown to reduce the expression of Foxo1 and contents of superoxide dismutase, catalase and glutathione peroxidase, yet increase lipocalin-2 expression as well as hydroxyproline and malondialdehyde contents. Also, Foxo1-mediated lipocalin-2 inhibition attenuated AD. Conclusion: Our study shows downregulating miR-96-5p limited AD progression, highlighting miR-96-5p a potential therapeutic target in treating AD.
Collapse
Affiliation(s)
- Bo-Wen Wu
- Department of Biochemistry, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, PR China.,Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang 050091, Hebei Province, PR China
| | - Jin-Dong Guo
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang 050091, Hebei Province, PR China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, PR China
| | - Mi-Shan Wu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang 050091, Hebei Province, PR China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, PR China
| | - Yu Liu
- Department of Biochemistry, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, PR China.,Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang 050091, Hebei Province, PR China
| | - Meng Lu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang 050091, Hebei Province, PR China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, PR China
| | - Yu-Hui Zhou
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang 050091, Hebei Province, PR China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, PR China
| | - Hong-Wei Han
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang 050091, Hebei Province, PR China.,Department of Formulaology, Basic Medicine College, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, PR China
| |
Collapse
|
33
|
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: 32] [Impact Index Per Article: 6.4] [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
|
34
|
Peters LJF, Floege J, Biessen EAL, Jankowski J, van der Vorst EPC. MicroRNAs in Chronic Kidney Disease: Four Candidates for Clinical Application. Int J Mol Sci 2020; 21:6547. [PMID: 32906849 PMCID: PMC7555601 DOI: 10.3390/ijms21186547] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
There are still major challenges regarding the early diagnosis and treatment of chronic kidney disease (CKD), which is in part due to the fact that its pathophysiology is very complex and not clarified in detail. The diagnosis of CKD commonly is made after kidney damage has occurred. This highlights the need for better mechanistic insight into CKD as well as improved clinical tools for both diagnosis and treatment. In the last decade, many studies have focused on microRNAs (miRs) as novel diagnostic tools or clinical targets. MiRs are small non-coding RNA molecules that are involved in post-transcriptional gene regulation and many have been studied in CKD. A wide array of pre-clinical and clinical studies have highlighted the potential role for miRs in the pathogenesis of hypertensive nephropathy, diabetic nephropathy, glomerulonephritis, kidney tubulointerstitial fibrosis, and some of the associated cardiovascular complications. In this review, we will provide an overview of the miRs studied in CKD, especially highlighting miR-103a-3p, miR-192-5p, the miR-29 family and miR-21-5p as these have the greatest potential to result in novel therapeutic and diagnostic strategies.
Collapse
Affiliation(s)
- Linsey J. F. Peters
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, 52074 Aachen, Germany; (L.J.F.P.); (E.A.L.B.); (J.J.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University Hospital, 52074 Aachen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, 52074 Aachen, Germany;
| | - Erik A. L. Biessen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, 52074 Aachen, Germany; (L.J.F.P.); (E.A.L.B.); (J.J.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, 52074 Aachen, Germany; (L.J.F.P.); (E.A.L.B.); (J.J.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, 52074 Aachen, Germany; (L.J.F.P.); (E.A.L.B.); (J.J.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University Hospital, 52074 Aachen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| |
Collapse
|
35
|
Upregulation of miR-382 contributes to renal fibrosis secondary to aristolochic acid-induced kidney injury via PTEN signaling pathway. Cell Death Dis 2020; 11:620. [PMID: 32796834 PMCID: PMC7429500 DOI: 10.1038/s41419-020-02876-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022]
Abstract
Acute kidney injury (AKI) has a critical role in the development of chronic kidney disease (CKD). Building on our previous findings, we explored the role of miR-382 in facilitating the transition of AKI to CKD using the Aristolochic acid (AA) nephropathy model, which was induced by intraperitoneal injection of aristolochic acid I salt (10 or 20 mg/kg). The effects of genetic depletion, pharmacologic inhibition, or overexpression of miR-382 on the PTEN/AKT signaling pathway were examined in vivo and in vitro. Changes in renal pathology and renal epithelial polarity were evaluated. A luciferase reporter assay was performed to investigate the reciprocal suppression relationship between miR-382 and PTEN. Renal fibrosis developed 14 d after AA exposure in a dose- and time-dependent manner. Renal abundance of miR-382 was upregulated following AA treatment, while genetic depletion or pharmacological inhibition of miR-382 partially reversed renal tubulointerstitial fibrosis. Expression of PTEN, a target of miR-382, was downregulated and subsequently its downstream AKT signaling pathway was activated during AKI to CKD transition induced by AA. Inhibition of PTEN in vitro resulted in the acquisition of the EMT phenotypes. Furthermore, upregulation of miR-382 in renal epithelial cells was partially mediated by the activation of NF-kB signaling, with a substantial elevation of proinflammatory cytokines. An in vivo study revealed that either miR-382 knockdown or miR-382 knockout was pivotal for inflammatory suppression, while an in vitro experiment confirmed that upregulation of miR-382 in cultured MTEC cells under AA exposure was remarkably reversed by NF-kB siRNA. These data indicated a novel role for the NF-κB/miR-382/PTEN/AKT axis in the pathogenesis of tubulointerstitial fibrosis following AA-induced acute renal tubular epithelial injury. Targeting miR-382 may lead to a potential novel therapeutic approach for retarding the AKT to CKD transition.
Collapse
|
36
|
Chao C, Yeh H, Han D, Huang J, Huang K. Determinants of circulating microRNA-125b, a risk predictor of vascular calcification, among community-dwelling older adults . Clin Transl Med 2020; 10:e145. [PMID: 32898327 PMCID: PMC7423181 DOI: 10.1002/ctm2.145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Chia‐Ter Chao
- Nephrology DivisionDepartment of Internal MedicineNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
- Graduate Institute of ToxicologyNational Taiwan University College of MedicineTaipeiTaiwan
- Geriatric and Community Medicine Research CenterNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
| | | | - Der‐Sheng Han
- Geriatric and Community Medicine Research CenterNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
- Department of Rehabilitation and Physical MedicineNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
| | - Jenq‐Wen Huang
- Nephrology DivisionDepartment of Internal MedicineNational Taiwan University Hospital Yunlin BranchYunlin CountyTaiwan
| | - Kuo‐Chin Huang
- Geriatric and Community Medicine Research CenterNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
- Department of Family MedicineNational Taiwan University Hospital BeiHu BranchTaipeiTaiwan
- Department of Family MedicineNational Taiwan University HospitalTaipeiTaiwan
- Department of Family MedicineCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
37
|
Roumeliotis S, Mallamaci F, Zoccali C. Endothelial Dysfunction in Chronic Kidney Disease, from Biology to Clinical Outcomes: A 2020 Update. J Clin Med 2020; 9:jcm9082359. [PMID: 32718053 PMCID: PMC7465707 DOI: 10.3390/jcm9082359] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
The vascular endothelium is a dynamic, functionally complex organ, modulating multiple biological processes, including vascular tone and permeability, inflammatory responses, thrombosis, and angiogenesis. Endothelial dysfunction is a threat to the integrity of the vascular system, and it is pivotal in the pathogenesis of atherosclerosis and cardiovascular disease. Reduced nitric oxide (NO) bioavailability is a hallmark of chronic kidney disease (CKD), with this disturbance being almost universal in patients who reach the most advanced phase of CKD, end-stage kidney disease (ESKD). Low NO bioavailability in CKD depends on several mechanisms affecting the expression and the activity of endothelial NO synthase (eNOS). Accumulation of endogenous inhibitors of eNOS, inflammation and oxidative stress, advanced glycosylation products (AGEs), bone mineral balance disorders encompassing hyperphosphatemia, high levels of the phosphaturic hormone fibroblast growth factor 23 (FGF23), and low levels of the active form of vitamin D (1,25 vitamin D) and the anti-ageing vasculoprotective factor Klotho all impinge upon NO bioavailability and are critical to endothelial dysfunction in CKD. Wide-ranging multivariate interventions are needed to counter endothelial dysfunction in CKD, an alteration triggering arterial disease and cardiovascular complications in this high-risk population.
Collapse
Affiliation(s)
- Stefanos Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, School of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Francesca Mallamaci
- CNR-IFC (National Research Council of Italy, Centre of Clinical Physiology, Clinical Epidemiology of Renal Diseases and Hypertension Unit, Reggio Cal., c/o Ospedali Riuniti, 89124 Reggio Cal, Italy;
| | - Carmine Zoccali
- CNR-IFC (National Research Council of Italy, Centre of Clinical Physiology, Clinical Epidemiology of Renal Diseases and Hypertension Unit, Reggio Cal., c/o Ospedali Riuniti, 89124 Reggio Cal, Italy;
- Correspondence: ; Tel.: +39-340-73540-62
| |
Collapse
|
38
|
Chen SI, Chiang CL, Chao CT, Chiang CK, Huang JW. Gustatory Function and the Uremic Toxin, Phosphate, Are Modulators of the Risk of Vascular Calcification among Patients with Chronic Kidney Disease: A Pilot Study. Toxins (Basel) 2020; 12:toxins12060420. [PMID: 32630499 PMCID: PMC7354456 DOI: 10.3390/toxins12060420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 12/25/2022] Open
Abstract
Patients with chronic kidney disease (CKD) have an increased risk of vascular calcification (VC), including aortic arch calcification (AAC). Few investigated the influence of gustatory function on the probability of having VC. We examined whether gustatory function results modulated the probability of having VC in patients with CKD. We prospectively enrolled adults with CKD (estimated glomerular filtration rate <60 mL/min/1.73 m2), with their AAC rated semi-quantitatively and gustatory function assessed by objective and subjective approaches. Multiple logistic regression was used to analyze the relationship between gustatory function results and AAC. Those with AAC had significantly better objective gustatory function in aggregate scores (p = 0.039) and categories (p = 0.022) and less defective bitter taste (p = 0.045) and scores (p = 0.037) than those without. Multiple regression analyses showed that higher aggregate scores (odds ratio (OR) 1.288, p = 0.032), or better gustatory function, and higher bitter taste scores (OR 2.558, p = 0.019) were each associated with a higher probability of having AAC among CKD patients; such an association was modulated by serum phosphate levels. In conclusion, better gustatory function was independently correlated with having AAC among CKD patients. A follow-up of VC severity may be an underrecognized component of care for CKD patients with a preserved gustatory function.
Collapse
Affiliation(s)
- Shih-I Chen
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital Beihu Branch, Taipei 108, Taiwan;
- Geriatric and Community Medicine Research Center, National Taiwan University Hospital BeiHu Branch, Taipei 108, Taiwan
| | - Chin-Ling Chiang
- Department of Nursing, National Taiwan University Hospital Beihu Branch, Taipei 108, Taiwan;
| | - Chia-Ter Chao
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital Beihu Branch, Taipei 108, Taiwan;
- Geriatric and Community Medicine Research Center, National Taiwan University Hospital BeiHu Branch, Taipei 108, Taiwan
- Graduate Institute of Toxicology, National Taiwan University, Taipei 10617, Taiwan;
- Correspondence: Chia-Ter Chao, ; Tel.: +886-2-23717101-5307; Fax: +886-2-23123456
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, National Taiwan University, Taipei 10617, Taiwan;
- Department of Integrative Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Jenq-Wen Huang
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin County 260, Taiwan;
| |
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: 22] [Impact Index Per Article: 4.4] [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
|
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: 6.4] [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
|
41
|
Yeh HY, Chao CT, Lai YP, Chen HW. Predicting the Associations between Meridians and Chinese Traditional Medicine Using a Cost-Sensitive Graph Convolutional Neural Network. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17030740. [PMID: 31979314 PMCID: PMC7036907 DOI: 10.3390/ijerph17030740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 12/17/2022]
Abstract
Natural products are the most important and commonly used in Traditional Chinese Medicine (TCM) for healthcare and disease prevention in East-Asia. Although the Meridian system of TCM was established several thousand years ago, the rationale of Meridian classification based on the ingredient compounds remains poorly understood. A core challenge for the traditional machine learning approaches for chemical activity prediction is to encode molecules into fixed length vectors but ignore the structural information of the chemical compound. Therefore, we apply a cost-sensitive graph convolutional neural network model to learn local and global topological features of chemical compounds, and discover the associations between TCM and their Meridians. In the experiments, we find that the performance of our approach with the area under the receiver operating characteristic curve (ROC-AUC) of 0.82 which is better than the traditional machine learning algorithm and also obtains 8%–13% improvement comparing with the state-of-the-art methods. We investigate the powerful ability of deep learning approach to learn the proper molecular descriptors for Meridian prediction and to provide novel insights into the complementary and alternative medicine of TCM.
Collapse
Affiliation(s)
- Hsiang-Yuan Yeh
- School of Big Data Management, Soochow University, Taipei 111, Taiwan
- Correspondence:
| | - Chia-Ter Chao
- Department of Medicine, National Taiwan University Hospital BeiHu Branch, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Yi-Pei Lai
- School of Big Data Management, Soochow University, Taipei 111, Taiwan
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
42
|
Chao CT, Yuan TH, Yeh HY, Chen HY, Huang JW, Chen HW. Risk Factors Associated With Altered Circulating Micro RNA -125b and Their Influences on Uremic Vascular Calcification Among Patients With End-Stage Renal Disease. J Am Heart Assoc 2020; 8:e010805. [PMID: 30646802 PMCID: PMC6497364 DOI: 10.1161/jaha.118.010805] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background MicroRNA‐125b (miR‐125b) has been shown to regulate vascular calcification (VC), and serum miR‐125b levels are a potential biomarker for estimating the risk of uremic VC status. However, it is unknown whether clinical features, including chronic kidney disease–mineral bone disorder molecules, affect serum miR‐125b levels. Methods and Results Patients receiving chronic dialysis for ≥3 months were recruited from different institutes. Serum miR‐125b and chronic kidney disease–mineral bone disorder effectors, including intact parathyroid hormone, 25‐OH‐D, fibroblast growth factor‐23, osteoprotegerin, and fetuin‐A, were quantified. We used multivariate regression analyses to identify factors associated with low serum miR‐125b levels and an area under receiver operating characteristic curve curve to derive optimal cutoffs for factors exhibiting close associations. Further regression analyses evaluated the influence of miR‐125b on VC risk. Among 223 patients receiving chronic dialysis (mean age, 67.3 years; mean years of dialysis, 5.2), 54 (24.2%) had high serum miR‐125b levels. Osteoprotegerin (P=0.013), fibroblast growth factor‐23 (P=0.006), and fetuin‐A (P=0.036) were linearly associated with serum miR‐125b levels. High osteoprotegerin levels independently correlated with high serum miR‐125 levels. Adding serum miR‐125b levels and serum osteoprotegerin levels (≥400 pg/mL) into models estimating the risk of uremic VC increased the area under receiver operating characteristic curve values (for models without miR‐125b/osteoprotegerin, with miR‐125b, and both: 0.74, 0.79, and 0.81, respectively). Conclusions Serum osteoprotegerin levels ≥400 pg/mL and serum miR‐125b levels synergistically increased the accuracy of estimating VC risk among patients receiving chronic dialysis. Taking miR‐125b and osteoprotegerin levels into consideration when estimating VC risk may be recommended.
Collapse
Affiliation(s)
- Chia-Ter Chao
- 1 Department of Medicine National Taiwan University Hospital BeiHu Branch Taipei Taiwan.,2 Nephrology Division Department of Internal Medicine National Taiwan University Hospital Taipei Taiwan.,3 Department of Geriatric and Community Medicine Research Center National Taiwan University Hospital BeiHu branch Taipei Taiwan
| | - Tzu-Hang Yuan
- 4 Graduate Institute of Toxicology National Taiwan University Taipei Taiwan
| | - Hsiang-Yuan Yeh
- 5 School of Big Data Management Soochow University Taipei Taiwan
| | - Hsuan-Yu Chen
- 6 Institute of Statistical Science Academia Sinica Taipei Taiwan
| | - Jenq-Wen Huang
- 2 Nephrology Division Department of Internal Medicine National Taiwan University Hospital Taipei Taiwan
| | - Huei-Wen Chen
- 4 Graduate Institute of Toxicology National Taiwan University Taipei Taiwan
| |
Collapse
|
43
|
MESH Headings
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/physiopathology
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Gene Expression Regulation
- Humans
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription, Genetic
- Vascular Diseases/genetics
- Vascular Diseases/pathology
- Vascular Diseases/physiopathology
- Vascular Remodeling/physiology
Collapse
Affiliation(s)
- Ning Shi
- Department of Surgery, University of Missouri, Columbia, MO
- Department of Physiology & pharmacology, The University of Georgia, Athens, GA
| | - Xiaohan Mei
- Department of Physiology & pharmacology, The University of Georgia, Athens, GA
| | - Shi-You Chen
- Department of Surgery, University of Missouri, Columbia, MO
- Department of Physiology & pharmacology, The University of Georgia, Athens, GA
- Correspondence to: Shi-You Chen, PhD, Department of Surgery, University of Missouri, 1 Hospital Drive, Columbia, MO 65212, , Tel: (573) 882-3137, Fax: (573)884-4585
| |
Collapse
|
44
|
Chao CT, Yeh HY, Tsai YT, Chuang PH, Yuan TH, Huang JW, Chen HW. Natural and non-natural antioxidative compounds: potential candidates for treatment of vascular calcification. Cell Death Discov 2019; 5:145. [PMID: 31754473 PMCID: PMC6853969 DOI: 10.1038/s41420-019-0225-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/23/2019] [Accepted: 09/27/2019] [Indexed: 12/18/2022] Open
Abstract
Vascular calcification (VC) is highly prevalent in patients with advanced age, or those with chronic kidney disease and diabetes, accounting for substantial global cardiovascular burden. The pathophysiology of VC involves active mineral deposition by transdifferentiated vascular smooth muscle cells exhibiting osteoblast-like behavior, building upon cores with or without apoptotic bodies. Oxidative stress drives the progression of the cellular phenotypic switch and calcium deposition in the vascular wall. In this review, we discuss potential compounds that shield these cells from the detrimental influences of reactive oxygen species as promising treatment options for VC. A comprehensive summary of the current literature regarding antioxidants for VC is important, as no effective therapy is currently available for this disease. We systematically searched through the existing literature to identify original articles investigating traditional antioxidants and novel compounds with antioxidant properties with regard to their effectiveness against VC in experimental or clinical settings. We uncovered 36 compounds with antioxidant properties against VC pathology, involving mechanisms such as suppression of NADPH oxidase, BMP-2, and Wnt/β-catenin; anti-inflammation; and activation of Nrf2 pathways. Only two compounds have been tested clinically. These findings suggest that a considerable opportunity exists to harness these antioxidants for therapeutic use for VC. In order to achieve this goal, more translational studies are needed.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Department of Medicine, National Taiwan University Hospital BeiHu Branch, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsiang-Yuan Yeh
- School of Big Data Management, Soochow University, Taipei, Taiwan
| | - You-Tien Tsai
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Huan Chuang
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tzu-Hang Yuan
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jenq-Wen Huang
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
45
|
Disthabanchong S, Srisuwarn P. Mechanisms of Vascular Calcification in Kidney Disease. Adv Chronic Kidney Dis 2019; 26:417-426. [PMID: 31831120 DOI: 10.1053/j.ackd.2019.08.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/18/2019] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
The increase in prevalence and severity of vascular calcification in chronic kidney disease is a result of complex interactions between changes in the vascular bed, mineral metabolites, and other uremic factors. Vascular calcification can occur in the intima and the media of arterial wall. Under permissive conditions, vascular smooth muscle cells (VSMCs) can transform to osteoblast-like phenotype. The membrane-bound vesicles released from transformed VSMCs and the apoptotic bodies derived from dying VSMCs serve as nucleating structures for calcium crystal formation. Alterations in the quality and the quantity of endogenous calcification inhibitors also give rise to an environment that potentiates calcification.
Collapse
Affiliation(s)
- Sinee Disthabanchong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Praopilad Srisuwarn
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
46
|
Valdivielso JM, Rodríguez-Puyol D, Pascual J, Barrios C, Bermúdez-López M, Sánchez-Niño MD, Pérez-Fernández M, Ortiz A. Atherosclerosis in Chronic Kidney Disease: More, Less, or Just Different? Arterioscler Thromb Vasc Biol 2019; 39:1938-1966. [PMID: 31412740 DOI: 10.1161/atvbaha.119.312705] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Patients with chronic kidney disease (CKD) are at an increased risk of premature mortality, mainly from cardiovascular causes. The association between CKD on hemodialysis and accelerated atherosclerosis was described >40 years ago. However, more recently, it has been suggested that the increase in atherosclerosis risk is actually observed in early CKD stages, remaining stable thereafter. In this regard, interventions targeting the pathogenesis of atherosclerosis, such as statins, successful in the general population, have failed to benefit patients with very advanced CKD. This raises the issue of the relative contribution of atherosclerosis versus other forms of cardiovascular injury such as arteriosclerosis or myocardial injury to the increased cardiovascular risk in CKD. In this review, the pathophysiogical contributors to atherosclerosis in CKD that are shared with the general population, or specific to CKD, are discussed. The NEFRONA study (Observatorio Nacional de Atherosclerosis en NEFrologia) prospectively assessed the prevalence and progression of subclinical atherosclerosis (plaque in vascular ultrasound), confirming an increased prevalence of atherosclerosis in patients with moderate CKD. However, the adjusted odds ratio for subclinical atherosclerosis increased with CKD stage, suggesting a contribution of CKD itself to subclinical atherosclerosis. Progression of atherosclerosis was closely related to CKD progression as well as to the baseline presence of atheroma plaque, and to higher phosphate, uric acid, and ferritin and lower 25(OH) vitamin D levels. These insights may help design future clinical trials of stratified personalized medicine targeting atherosclerosis in patients with CKD. Future primary prevention trials should enroll patients with evidence of subclinical atherosclerosis and should provide a comprehensive control of all known risk factors in addition to testing any additional intervention or placebo.
Collapse
Affiliation(s)
- José M Valdivielso
- From the Vascular & Renal Translational Research Group and UDETMA, IRBLleida. Spanish Research Network for Renal Diseases (RedInRen. ISCIII), Lleida, Spain (J.M.V., M.B.-L.)
| | - Diego Rodríguez-Puyol
- Nephrology Unit, Fundación para la investigación del Hospital Universitario Príncipe de Asturias, RedInRen, Alcalá de Henares, Madrid, Spain (D.R.-P.)
| | - Julio Pascual
- Department of Nephrology, Institute Mar for Medical Research, Hospital del Mar, RedInRen, Barcelona, Spain (J.P., C.B.)
| | - Clara Barrios
- Department of Nephrology, Institute Mar for Medical Research, Hospital del Mar, RedInRen, Barcelona, Spain (J.P., C.B.)
| | - Marcelino Bermúdez-López
- From the Vascular & Renal Translational Research Group and UDETMA, IRBLleida. Spanish Research Network for Renal Diseases (RedInRen. ISCIII), Lleida, Spain (J.M.V., M.B.-L.)
| | - Maria Dolores Sánchez-Niño
- IIS-Fundacion Jimenez Diaz, School of Medicine, University Autonoma of Madrid, FRIAT and RedInRen, Madrid, Spain (M.D.S.-N., A.O.)
| | | | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, School of Medicine, University Autonoma of Madrid, FRIAT and RedInRen, Madrid, Spain (M.D.S.-N., A.O.)
| |
Collapse
|
47
|
Lu Y, Thavarajah T, Gu W, Cai J, Xu Q. Impact of miRNA in Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 38:e159-e170. [PMID: 30354259 DOI: 10.1161/atvbaha.118.310227] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yao Lu
- From the Center of Clinical Pharmacology (Y.L.)
| | - Tanuja Thavarajah
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Wenduo Gu
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Jingjing Cai
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingbo Xu
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| |
Collapse
|
48
|
Chao CT, Yeh HY, Yuan TH, Chiang CK, Chen HW. MicroRNA-125b in vascular diseases: An updated systematic review of pathogenetic implications and clinical applications. J Cell Mol Med 2019; 23:5884-5894. [PMID: 31301111 PMCID: PMC6714222 DOI: 10.1111/jcmm.14535] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/02/2019] [Accepted: 06/19/2019] [Indexed: 12/26/2022] Open
Abstract
Epigenetic changes, particularly non‐coding RNAs, have been implicated extensively in the pathogenesis of vascular diseases. Specific miRNAs are involved in the differentiation, phenotypic switch, proliferation, apoptosis, cytokine production and matrix deposition of endothelial cells and/or vascular smooth muscle cells. MicroRNA‐125b has been studied in depth for its role in carcinogenesis with a double‐edged role; that is, it can act as an oncogene in some cancer types and as a tumour suppressor gene in others. However, cumulative evidence from the use of advanced miRNA profiling techniques and bioinformatics analysis suggests that miR‐125b can be a potential mediator and useful marker of vascular diseases. Currently, the exact role of miR‐125b in vascular diseases is not known. In this systematic review, we intend to provide an updated compilation of all the recent findings of miR‐125b in vascular diseases, using a systematic approach of retrieving data from all available reports followed by data summarization. MiR‐125b serves as a pathogenic player in multiple vascular pathologies involving endothelia and vascular smooth muscle cells and also serves as a diagnostic marker for vascular diseases. We further provide a computational biologic presentation of the complex network of miR‐125b and its target genes within the scope of vascular diseases.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Department of Medicine, National Taiwan University Hospital BeiHu Branch, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsiang-Yuan Yeh
- School of Big Data Management, Soochow University, Taipei, Taiwan
| | - Tzu-Hang Yuan
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
49
|
Lee CT, Lee YT, Tain YL, Ng HY, Kuo WH. Circulating microRNAs and vascular calcification in hemodialysis patients. J Int Med Res 2019; 47:2929-2939. [PMID: 31144545 PMCID: PMC6683928 DOI: 10.1177/0300060519848949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/16/2019] [Indexed: 12/16/2022] Open
Abstract
Objective Vascular calcification is common in chronic dialysis patients and is associated with increased morbidity and mortality. However, the role of circulating microRNAs (miRs) in vascular calcification has rarely been investigated. We aimed to determine circulating levels of miRs in hemodialysis patients, and analyzed their relationship with vascular calcification. Methods Sixty-one stable hemodialysis patients were enrolled, including 31 with vascular calcification and 30 without. Demographic and biochemical data were collected and reviewed. The presence and severity of vascular calcification were determined by lumber spine X-ray. Blood levels of miR29a/b, miR223, miR9, and miR21 were determined. Results Patients with vascular calcification were older (65.6 ± 9.0 vs. 59.1 ± 7.1 years) with a higher proportion of vascular disease (55% vs. 23%) than those without vascular calcification. Additionally, high-sensitivity C-reactive protein (3.90 vs 2.09 mg/dL) and fibroblast growth factor 23 (17311 vs. 6306 pg/mL) were significantly higher. Patients with vascular calcification also had higher levels of miR29a/b and miR223. Regression analysis indicated that age and miR29a were significant associates of the calcification score. Conclusions Hemodialysis patients with vascular calcification had higher levels of miR 29a/b and miR223 than those without vascular calcification, and circulating miR29a was associated with calcification severity.
Collapse
Affiliation(s)
- Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yueh-Ting Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - You-Lin Tain
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hwee-Yeong Ng
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wei-Hung Kuo
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
50
|
Su Y, Li Q, Zheng Z, Wei X, Hou P. Integrative bioinformatics analysis of miRNA and mRNA expression profiles and identification of associated miRNA-mRNA network in aortic dissection. Medicine (Baltimore) 2019; 98:e16013. [PMID: 31192949 PMCID: PMC6587623 DOI: 10.1097/md.0000000000016013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Aortic dissection (AD) is one of the most lethal cardiovascular diseases. The aim of this study was to identify core genes and pathways revealing pathogenesis in AD. METHODS We screened differentially expressed mRNAs and miRNAs using mRNA and miRNA expression profile data of AD from Gene Expression Omnibus. Then functional and pathway enrichment analyses of differential expression genes (DEGs) was performed utilizing the database for annotation, visualization, and integrated discovery (DAVID). Target genes with differential expression miRNAs (DEMIs) were predicted using the miRWalk database, and the intersection between these predictions and DEGs was selected as differentially expressed miRNA-target genes. In addition, a protein-protein interaction (PPI) network and miRNA-mRNA regulatory network were constructed. RESULTS In total, 130 DEGs and 47 DEMIs were identified from mRNA and miRNA microarray, respectively, and 45 DEGs were DEMI-target genes. The PPI and miRNA-mRNA network included 79 node genes and 74 node genes, respectively, while 23 hub genes and 2 hub miRNAs were identified. The DEGs, PPI and modules differential expression miRNA-target genes were all mainly enriched in cell cycle, cell proliferation and cell apoptosis signaling pathways. CONCLUSION Taken above, the study reveals some candidate genes and pathways potentially involving molecular mechanisms of AD. These findings provide a new insight for research and treatment of AD.
Collapse
|