1
|
Cui X, Wei W, Hu Y, Zhang Z, Lu M, Li Y, Wu J, Li C. Dietary inflammation and vascular calcification: a comprehensive review of the associations, underlying mechanisms, and prevention strategies. Crit Rev Food Sci Nutr 2024:1-22. [PMID: 39340196 DOI: 10.1080/10408398.2024.2408447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2024]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death globally, and vascular calcification (VC) has been recognized as an independent and strong predictor of global CVD and mortality. Chronic inflammation has been demonstrated to play a significant role in the progression of VC. This review aims to summarize the literature that aimed to elucidate the associations between dietary inflammation (DI) and VC as well as to explore the mechanisms underlying the association and discuss strategies (including dietary interventions) to prevent VC. Notably, diets rich in processed foods, carbohydrates with high glycemic index/load, saturated fatty acids, trans-fatty acids, cholesterol, and phosphorus were found to induce inflammatory responses and accelerate the progression of VC, indicating a close relationship between DI and VC. Moreover, we demonstrate that an imbalance in the composition of the gut microbiota caused by the intake of specific dietary choices favored the production of certain metabolites that may contribute to the progression of VC. The release of inflammatory and adhesion cytokines, activation of inflammatory pathways, oxidative stress, and metabolic disorders were noted to be the main mechanisms through which DI induced VC. To reduce and slow the progression of VC, emphasis should be placed on the intake of diets rich in omega-3 fatty acids, dietary fiber, Mg, Zn, and polyphenols, as well as the adjustment of dietary pattern to reduce the risk of VC. This review is expected to be useful for guiding future research on the interplay between DI and VC.
Collapse
Affiliation(s)
- Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wendi Wei
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
2
|
McCallion S, McLarnon T, Cooper E, English AR, Watterson S, Chemaly ME, McGeough C, Eakin A, Ahmed T, Gardiner P, Pendleton A, Wright G, McGuigan D, O’Kane M, Peace A, Kuan Y, Gibson DS, McClean PL, Kelly C, McGilligan V, Murray EK, McCarroll F, Bjourson AJ, Rai TS. Senescence Biomarkers CKAP4 and PTX3 Stratify Severe Kidney Disease Patients. Cells 2024; 13:1613. [PMID: 39404377 PMCID: PMC11475272 DOI: 10.3390/cells13191613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
INTRODUCTION Cellular senescence is the irreversible growth arrest subsequent to oncogenic mutations, DNA damage, or metabolic insult. Senescence is associated with ageing and chronic age associated diseases such as cardiovascular disease and diabetes. The involvement of cellular senescence in acute kidney injury (AKI) and chronic kidney disease (CKD) is not fully understood. However, recent studies suggest that such patients have a higher-than-normal level of cellular senescence and accelerated ageing. METHODS This study aimed to discover key biomarkers of senescence in AKI and CKD patients compared to other chronic ageing diseases in controls using OLINK proteomics. RESULTS We show that senescence proteins CKAP4 (p-value < 0.0001) and PTX3 (p-value < 0.0001) are upregulated in AKI and CKD patients compared with controls with chronic diseases, suggesting the proteins may play a role in overall kidney disease development. CONCLUSIONS CKAP4 was found to be differentially expressed in both AKI and CKD when compared to UHCs; hence, this biomarker could be a prognostic senescence biomarker of both AKI and CKD.
Collapse
Affiliation(s)
- Sean McCallion
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Thomas McLarnon
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Eamonn Cooper
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Andrew R. English
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
- School of Health and Life Sciences, Teesside University, Campus Heart, Middlesbrough TS1 3BX, UK
| | - Steven Watterson
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Melody El Chemaly
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Cathy McGeough
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Amanda Eakin
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Tan Ahmed
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Philip Gardiner
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - Adrian Pendleton
- Belfast Health and Social Care Trust (BHSCT), Belfast City Hospital, Belfast BT9 7AB, UK
| | - Gary Wright
- Belfast Health and Social Care Trust (BHSCT), Belfast City Hospital, Belfast BT9 7AB, UK
| | - Declan McGuigan
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Maurice O’Kane
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - Aaron Peace
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - Ying Kuan
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - David S. Gibson
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Paula L. McClean
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Catriona Kelly
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Victoria McGilligan
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Elaine K. Murray
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Frank McCarroll
- Western Health and Social Care Trust (WHSCT), Altnagelvin Area Hospital, Londonderry BT47 6SB, UK
| | - Anthony J. Bjourson
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| | - Taranjit Singh Rai
- Personalised Medicine Centre, School of Medicine, Ulster University, Londonderry BT48 7JL, UK
| |
Collapse
|
3
|
Ethier I, Hayat A, Pei J, Hawley CM, Johnson DW, Francis RS, Wong G, Craig JC, Viecelli AK, Htay H, Ng S, Leibowitz S, Cho Y. Peritoneal dialysis versus haemodialysis for people commencing dialysis. Cochrane Database Syst Rev 2024; 6:CD013800. [PMID: 38899545 PMCID: PMC11187793 DOI: 10.1002/14651858.cd013800.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
BACKGROUND Peritoneal dialysis (PD) and haemodialysis (HD) are two possible modalities for people with kidney failure commencing dialysis. Only a few randomised controlled trials (RCTs) have evaluated PD versus HD. The benefits and harms of the two modalities remain uncertain. This review includes both RCTs and non-randomised studies of interventions (NRSIs). OBJECTIVES To evaluate the benefits and harms of PD, compared to HD, in people with kidney failure initiating dialysis. SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies from 2000 to June 2024 using search terms relevant to this review. Studies in the Register were identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal, and ClinicalTrials.gov. MEDLINE and EMBASE were searched for NRSIs from 2000 until 28 March 2023. SELECTION CRITERIA RCTs and NRSIs evaluating PD compared to HD in people initiating dialysis were eligible. DATA COLLECTION AND ANALYSIS Two investigators independently assessed if the studies were eligible and then extracted data. Risk of bias was assessed using standard Cochrane methods, and relevant outcomes were extracted for each report. The primary outcome was residual kidney function (RKF). Secondary outcomes included all-cause, cardiovascular and infection-related death, infection, cardiovascular disease, hospitalisation, technique survival, life participation and fatigue. MAIN RESULTS A total of 153 reports of 84 studies (2 RCTs, 82 NRSIs) were included. Studies varied widely in design (small single-centre studies to international registry analyses) and in the included populations (broad inclusion criteria versus restricted to more specific participants). Additionally, treatment delivery (e.g. automated versus continuous ambulatory PD, HD with catheter versus arteriovenous fistula or graft, in-centre versus home HD) and duration of follow-up varied widely. The two included RCTs were deemed to be at high risk of bias in terms of blinding participants and personnel and blinding outcome assessment for outcomes pertaining to quality of life. However, most other criteria were assessed as low risk of bias for both studies. Although the risk of bias (Newcastle-Ottawa Scale) was generally low for most NRSIs, studies were at risk of selection bias and residual confounding due to the constraints of the observational study design. In children, there may be little or no difference between HD and PD on all-cause death (6 studies, 5752 participants: RR 0.81, 95% CI 0.62 to 1.07; I2 = 28%; low certainty) and cardiovascular death (3 studies, 7073 participants: RR 1.23, 95% CI 0.58 to 2.59; I2 = 29%; low certainty), and was unclear for infection-related death (4 studies, 7451 participants: RR 0.98, 95% CI 0.39 to 2.46; I2 = 56%; very low certainty). In adults, compared with HD, PD had an uncertain effect on RKF (mL/min/1.73 m2) at six months (2 studies, 146 participants: MD 0.90, 95% CI 0.23 to 3.60; I2 = 82%; very low certainty), 12 months (3 studies, 606 participants: MD 1.21, 95% CI -0.01 to 2.43; I2 = 81%; very low certainty) and 24 months (3 studies, 334 participants: MD 0.71, 95% CI -0.02 to 1.48; I2 = 72%; very low certainty). PD had uncertain effects on residual urine volume at 12 months (3 studies, 253 participants: MD 344.10 mL/day, 95% CI 168.70 to 519.49; I2 = 69%; very low certainty). PD may reduce the risk of RKF loss (3 studies, 2834 participants: RR 0.55, 95% CI 0.44 to 0.68; I2 = 17%; low certainty). Compared with HD, PD had uncertain effects on all-cause death (42 studies, 700,093 participants: RR 0.87, 95% CI 0.77 to 0.98; I2 = 99%; very low certainty). In an analysis restricted to RCTs, PD may reduce the risk of all-cause death (2 studies, 1120 participants: RR 0.53, 95% CI 0.32 to 0.86; I2 = 0%; moderate certainty). PD had uncertain effects on both cardiovascular (21 studies, 68,492 participants: RR 0.96, 95% CI 0.78 to 1.19; I2 = 92%) and infection-related death (17 studies, 116,333 participants: RR 0.90, 95% CI 0.57 to 1.42; I2 = 98%) (both very low certainty). Compared with HD, PD had uncertain effects on the number of patients experiencing bacteraemia/bloodstream infection (2 studies, 2582 participants: RR 0.34, 95% CI 0.10 to 1.18; I2 = 68%) and the number of patients experiencing infection episodes (3 studies, 277 participants: RR 1.23, 95% CI 0.93 to 1.62; I2 = 20%) (both very low certainty). PD may reduce the number of bacteraemia/bloodstream infection episodes (2 studies, 2637 participants: RR 0.44, 95% CI 0.27 to 0.71; I2 = 24%; low certainty). Compared with HD; It is uncertain whether PD reduces the risk of acute myocardial infarction (4 studies, 110,850 participants: RR 0.90, 95% CI 0.74 to 1.10; I2 = 55%), coronary artery disease (3 studies, 5826 participants: RR 0.95, 95% CI 0.46 to 1.97; I2 = 62%); ischaemic heart disease (2 studies, 58,374 participants: RR 0.86, 95% CI 0.57 to 1.28; I2 = 95%), congestive heart failure (3 studies, 49,511 participants: RR 1.10, 95% CI 0.54 to 2.21; I2 = 89%) and stroke (4 studies, 102,542 participants: RR 0.94, 95% CI 0.90 to 0.99; I2 = 0%) because of low to very low certainty evidence. Compared with HD, PD had uncertain effects on the number of patients experiencing hospitalisation (4 studies, 3282 participants: RR 0.90, 95% CI 0.62 to 1.30; I2 = 97%) and all-cause hospitalisation events (4 studies, 42,582 participants: RR 1.02, 95% CI 0.81 to 1.29; I2 = 91%) (very low certainty). None of the included studies reported specifically on life participation or fatigue. However, two studies evaluated employment. Compared with HD, PD had uncertain effects on employment at one year (2 studies, 593 participants: RR 0.83, 95% CI 0.20 to 3.43; I2 = 97%; very low certainty). AUTHORS' CONCLUSIONS The comparative effectiveness of PD and HD on the preservation of RKF, all-cause and cause-specific death risk, the incidence of bacteraemia, other vascular complications (e.g. stroke, cardiovascular events) and patient-reported outcomes (e.g. life participation and fatigue) are uncertain, based on data obtained mostly from NRSIs, as only two RCTs were included.
Collapse
Affiliation(s)
- Isabelle Ethier
- Department of Nephrology, Centre hospitalier de l'Université de Montréal, Montréal, Canada
- Health innovation and evaluation hub, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Canada
| | - Ashik Hayat
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Juan Pei
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
- Department of Nephrology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Carmel M Hawley
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - David W Johnson
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Ross S Francis
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Germaine Wong
- School of Public Health, The University of Sydney, Sydney, Australia
| | - Jonathan C Craig
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Andrea K Viecelli
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| | - Htay Htay
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | - Samantha Ng
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Saskia Leibowitz
- Department of Nephrology, Logan Hospital, Meadowbrook, Australia
| | - Yeoungjee Cho
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Australia
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, Australia
- Translational Research Institute, Brisbane, Australia
| |
Collapse
|
4
|
Miyazaki-Anzai S, Masuda M, Keenan AL, Shiozaki Y, Miranda JG, Miyazaki M. Activation of the IKK2/NF-κB pathway in VSMCs inhibits calcified vascular stiffness in CKD. JCI Insight 2024; 9:e174977. [PMID: 38470493 PMCID: PMC11128211 DOI: 10.1172/jci.insight.174977] [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: 08/25/2023] [Accepted: 02/29/2024] [Indexed: 03/13/2024] Open
Abstract
IKK2/NF-κB pathway-mediated inflammation in vascular smooth muscle cells (VSMCs) has been proposed to be an etiologic factor in medial calcification and stiffness. However, the role of the IKK2/NF-κB pathway in medial calcification remains to be elucidated. In this study, we found that chronic kidney disease (CKD) induces inflammatory pathways through the local activation of the IKK2/NF-κB pathway in VMSCs associated with calcified vascular stiffness. Despite reducing the expression of inflammatory mediators, complete inhibition of the IKK2/NF-κB pathway in vitro and in vivo unexpectedly exacerbated vascular mineralization and stiffness. In contrast, activation of NF-κB by SMC-specific IκBα deficiency attenuated calcified vascular stiffness in CKD. Inhibition of the IKK2/NF-κB pathway induced cell death of VSMCs by reducing anti-cell death gene expression, whereas activation of NF-κB reduced CKD-dependent vascular cell death. In addition, increased calcification of extracellular vesicles through the inhibition of the IKK2/NF-κB pathway induced mineralization of VSMCs, which was significantly reduced by blocking cell death in vitro and in vivo. This study reveals that activation of the IKK2/NF-κB pathway in VSMCs plays a protective role in CKD-dependent calcified vascular stiffness by reducing the release of apoptotic calcifying extracellular vesicles.
Collapse
|
5
|
Ding H, Zhu J, Tian Y, Xu L, Song L, Shi Y, Mu D, Chen R, Liu H, Liu B. Relationship between the triglyceride-glucose index and coronary artery calcification in asymptomatic, non-diabetic patients undergoing maintenance hemodialysis. Ren Fail 2023; 45:2200849. [PMID: 37133817 PMCID: PMC10158539 DOI: 10.1080/0886022x.2023.2200849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
OBJECTIVE Coronary artery calcification (CAC) is positively and independently associated with cardiovascular disease (CVD) in patients undergoing maintenance hemodialysis (MHD). Insulin resistance is independently associated with CAC and is an important risk factor for CVD. The triglyceride-glucose (TyG) index is a reliable biomarker of insulin resistance. This cross-sectional, observational study aimed to investigate the relationship between the TyG index and CAC in asymptomatic non-diabetic patients undergoing MHD. METHODS The quantitative coronary artery calcification score (CACS) was calculated and expressed using the Agatston score. The TyG index was calculated as ln [fasting triglyceride (mg/dL) × fasting glucose (mg/dL)/2]. Multiple Poisson regression analysis, Spearman correlation analysis, and receiver operating characteristic (ROC) curves were used to investigate the relationship between the TyG index and CAC. RESULTS The 151 patients were divided into three groups according to the tertiles of the TyG index. With an increase in the TyG index, the CACS significantly increased (Spearman's rho = 0.414, p < 0.001). Poisson regression analysis indicated that the TyG index was independently related to the presence of CAC (prevalence ratio, 1.281 [95% confidence interval, 1.121-1.465], p < 0.001). Furthermore, ROC curve analysis showed that the TyG index was of value in predicting the CAC in asymptomatic non-diabetic patients undergoing MHD, with an area under the curve of 0.667 (p = 0.010). CONCLUSION The TyG index is independently related to the presence of CAC in asymptomatic, non-diabetic patients undergoing MHD.
Collapse
Affiliation(s)
- Hong Ding
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Jinhua Zhu
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Ying Tian
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Li Xu
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Lei Song
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Ying Shi
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Dongxing Mu
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Ruoxin Chen
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Hong Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Bicheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
6
|
Dragoș D, Timofte D, Georgescu MT, Manea MM, Vacaroiu IA, Ionescu D, Balcangiu-Stroescu AE. Cardiovascular Calcifications Are Correlated with Inflammation in Hemodialysis Patients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1801. [PMID: 37893519 PMCID: PMC10608311 DOI: 10.3390/medicina59101801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: The main cause of morbidity and mortality in hemodialysis patients is cardiovascular disease, which is quite common. The main objective of our study was to investigate the relationship between oxidative stress, inflammation, and vascular and valvular calcifications in hemodialysis patients. Materials and Methods: This observational study had 54 hemodialysis patients, with an average age of 60.46 ± 13.18 years. Cardiovascular ultrasound was used to detect and/or measure aortic and mitral valve calcifications, carotid and femoral atheroma plaques, and common carotid intima-media thickness. The aortic calcification score was determined using a lateral abdomen plain radiograph. The inflammatory, oxidative, metabolic, and dietary statuses, as well as demographic characteristics, were identified. Results: There were significant correlations between the levels of IL-6 and carotid plaque number (p = 0.003), fibrinogen level and aortic valve calcifications (p = 0.05), intima-media thickness (p = 0.0007), carotid plaque number (p = 0.035), femoral plaque number (p = 0.00014), and aortic calcifications score (p = 0.0079). Aortic annulus calcifications (p = 0.03) and intima-media thickness (p = 0.038) were adversely linked with TNF-α. Nutrition parameters were negatively correlated with atherosclerosis markers: number of carotid plaques with albumin (p = 0.013), body mass index (p = 0.039), and triglycerides (p = 0.021); number of femoral plaques with phosphorus (0.013), aortic calcifications score with albumin (p = 0.051), intima-media thickness with LDL-cholesterol (p = 0.042). Age and the quantity of carotid plaques, femoral plaques, and aortic calcifications were linked with each other (p = 0.0022, 0.00011, and 0.036, respectively). Aortic annulus calcifications (p = 0.011), aortic valve calcifications (p = 0.023), and mitral valve calcifications (p = 0.018) were all associated with an increased risk of death. Conclusions: Imaging measures of atherosclerosis are adversely connected with dietary status and positively correlated with markers of inflammation and risk of mortality.
Collapse
Affiliation(s)
- Dorin Dragoș
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- 1st Internal Medicine Clinic, University Emergency Hospital Bucharest, Splaiul Independentei nr 169, Sect 5, 050098 Bucharest, Romania
| | - Delia Timofte
- Department of Dialysis, University Emergency Hospital Bucharest, Splaiul Independentei nr 168, Sect 5, 050098 Bucharest, Romania
| | - Mihai-Teodor Georgescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- Department of Radiotherapy, Prof. Dr. Al. Trestioreanu Institute of Oncology Bucharest, Șos. Fundeni nr 252, Sect 2, 022328 Bucharest, Romania
| | - Maria-Mirabela Manea
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- National Institute of Neurology and Cerebrovascular Diseases, Șos. Berceni nr 10–12, Sect 4, 041915 Bucharest, Romania
| | - Ileana Adela Vacaroiu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- Nephrology and Dialysis Clinic, “Sf. Ioan” Emergency Clinical Hospital, Șos. Vitan-Barzești nr 12, 042122 Bucharest, Romania
| | - Dorin Ionescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- Nephrology Clinic, University Emergency Hospital, Splaiul Independentei nr 169, Sect 5, 050098 Bucharest, Romania
| | - Andra-Elena Balcangiu-Stroescu
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania
| |
Collapse
|
7
|
Zhu E, Shu X, Xu Z, Peng Y, Xiang Y, Liu Y, Guan H, Zhong M, Li J, Zhang LZ, Nie R, Zheng Z. Screening of immune-related secretory proteins linking chronic kidney disease with calcific aortic valve disease based on comprehensive bioinformatics analysis and machine learning. J Transl Med 2023; 21:359. [PMID: 37264340 DOI: 10.1186/s12967-023-04171-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/30/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is one of the most significant cardiovascular risk factors, playing vital roles in various cardiovascular diseases such as calcific aortic valve disease (CAVD). We aim to explore the CKD-associated genes potentially involving CAVD pathogenesis, and to discover candidate biomarkers for the diagnosis of CKD with CAVD. METHODS Three CAVD, one CKD-PBMC and one CKD-Kidney datasets of expression profiles were obtained from the GEO database. Firstly, to detect CAVD key genes and CKD-associated secretory proteins, differentially expressed analysis and WGCNA were carried out. Protein-protein interaction (PPI), functional enrichment and cMAP analyses were employed to reveal CKD-related pathogenic genes and underlying mechanisms in CKD-related CAVD as well as the potential drugs for CAVD treatment. Then, machine learning algorithms including LASSO regression and random forest were adopted for screening candidate biomarkers and constructing diagnostic nomogram for predicting CKD-related CAVD. Moreover, ROC curve, calibration curve and decision curve analyses were applied to evaluate the diagnostic performance of nomogram. Finally, the CIBERSORT algorithm was used to explore immune cell infiltration in CAVD. RESULTS The integrated CAVD dataset identified 124 CAVD key genes by intersecting differential expression and WGCNA analyses. Totally 983 CKD-associated secretory proteins were screened by differential expression analysis of CKD-PBMC/Kidney datasets. PPI analysis identified two key modules containing 76 nodes, regarded as CKD-related pathogenic genes in CAVD, which were mostly enriched in inflammatory and immune regulation by enrichment analysis. The cMAP analysis exposed metyrapone as a more potential drug for CAVD treatment. 17 genes were overlapped between CAVD key genes and CKD-associated secretory proteins, and two hub genes were chosen as candidate biomarkers for developing nomogram with ideal diagnostic performance through machine learning. Furthermore, SLPI/MMP9 expression patterns were confirmed in our external cohort and the nomogram could serve as novel diagnosis models for distinguishing CAVD. Finally, immune cell infiltration results uncovered immune dysregulation in CAVD, and SLPI/MMP9 were significantly associated with invasive immune cells. CONCLUSIONS We revealed the inflammatory-immune pathways underlying CKD-related CAVD, and developed SLPI/MMP9-based CAVD diagnostic nomogram, which offered novel insights into future serum-based diagnosis and therapeutic intervention of CKD with CAVD.
Collapse
Affiliation(s)
- Enyi Zhu
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaorong Shu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zi Xu
- Department of Radiology, Guizhou Provincial People's Hospital, Guizhou, China
| | - Yanren Peng
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunxiu Xiang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yu Liu
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hui Guan
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ming Zhong
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jinhong Li
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Li-Zhen Zhang
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Ruqiong Nie
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
8
|
Ashour WM, Zamzam MSA, Sayed Ali HEEDE, Ebrahim RH. Effect of fetuin-A on adenine-induced chronic kidney disease model in male rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:511-516. [PMID: 37051102 PMCID: PMC10083833 DOI: 10.22038/ijbms.2023.66346.14584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 02/07/2023] [Indexed: 04/14/2023]
Abstract
Objectives This study aimed to investigate the possible effects of fetuin-A on an adenine-induced chronic kidney disease (CKD) model in male rats. Materials and Methods Rats were divided into three groups: group A included rats fed a normal diet; group B included rats fed a normal diet with 220 mg/kg adenine daily for 21 days; group C included rats fed a normal diet with 220 mg/kg adenine daily for 21 days and intraperitoneally administered with 5 mg\kg fetuin-A every other day for 2 weeks. Serum samples were assayed for serum creatinine, urea, sodium, potassium, calcium, phosphorus, tumor necrosis factor (TNF), interleukin-6 (IL-6), and estimated glomerular filtration rate (eGFR), and immunohistochemical staining was performed. Results Group B showed a significant increase in serum creatinine, urea, phosphorus, potassium, TNF, and IL-6 and a significant decrease in serum sodium, calcium, and eGFR compared with group A. Regarding immunohistochemistry, group B showed increased apoptosis. In group C, fetuin-A reduced the urea, creatinine, and phosphorus levels, and in group C, fetuin-A decreased inflammation and apoptosis by reduction of caspase-3 staining. Conclusion Fetuin-A improved kidney function in CKD due to its anti-inflammatory and anti-fibrotic role.
Collapse
Affiliation(s)
- Wesam M.R Ashour
- Physiology Department, Faculty of Medicine, Zagazig university, Sharqia government, Egypt
| | | | | | - Reham Hassan Ebrahim
- Physiology Department, Faculty of Medicine, Zagazig university, Sharqia government, Egypt
- Corresponding author: Reham Hassan Ebrahim. Physiology Department, Faculty of Medicine, Zagazig university, Sharqia government, Egypt.
| |
Collapse
|
9
|
Xiang X, He J, Zhang W, He Q, Liu Y. Coronary artery calcification in patients with advanced chronic kidney disease. BMC Cardiovasc Disord 2022; 22:453. [PMID: 36309659 PMCID: PMC9618197 DOI: 10.1186/s12872-022-02879-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction Cardiovascular disease (CVD) is associated with higher morbidity and mortality rates in patients with chronic kidney disease (CKD). Studies have shown that vascular calcification is a major predictor of CVD. Vascular calcification in the CKD population is associated with various risk factors, and changes in bone and mineral metabolism have been linked to an increased risk of atherosclerosis. Therefore, we aimed to investigate the correlation between vascular calcification and bone metabolism, which is necessary to improve the survival and prognosis of patients with CKD. Methods We included 146 patients with CKD who received coronary artery calcification (CAC) scores at our hospital from May 2017 to November 2018. Spearman rank correlation analysis, Mann–Whitney U test, and Kaplan–Meier method were used to analyze laboratory data and all-cause mortality. Results In the 146 patients, chronic glomerulonephritis accounted for the most common cause of CKD, at approximately 39.0%. Spearman rank correlation analysis on the factors influencing vascular calcification in patients with CKD showed that CAC score was significantly and positively correlated with C-reactive protein, N-terminal/midregion osteocalcin (N-MID), N-terminal peptide of type 1 procollagen (P1NP), β-cross-linked C-telopeptide of type 1 collagen (β-CTx), and parathyroid hormone (P = 0.0423, P = 0.0432, P = 0.0235, P = 0.0061, P < 0.0001, respectively). Serum calcium levels were positively correlated with N-MID, P1NP, β-CTx, and iPTH (r = 0.19, r = 0.24, r = 0.21, r = 0.21, respectively), and serum phosphorus levels were positively correlated with N-MID, P1NP, β-CTx, and iPTH (r = 0.50, r = 0.37, r = 0.50, r = 0.55, respectively). However, no difference was found in CVC scores among patients with CKD in different stages and receiving different treatments. In the Kaplan–Meier analysis of all-cause hospitalization and mortality rates, patients with CAC > 400 had a higher risk. Conclusion We found that the primary cause of CKD is glomerulonephritis, and the CAC score is positively correlated with inflammatory and bone metabolism markers, with a higher risk of all-cause mortality and cardiovascular hospitalization when the CAC score is greater than 400.
Collapse
|
10
|
Mas-Bargues C, Borrás C, Alique M. The Contribution of Extracellular Vesicles From Senescent Endothelial and Vascular Smooth Muscle Cells to Vascular Calcification. Front Cardiovasc Med 2022; 9:854726. [PMID: 35498012 PMCID: PMC9051028 DOI: 10.3389/fcvm.2022.854726] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/11/2022] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification is an irreversible pathological process associated with a loss of vascular wall function. This process occurs as a result of aging and age-related diseases, such as cardiovascular and chronic kidney diseases, and leads to comorbidities. During these age-related diseases, the endothelium accumulates senescent cells, which stimulate calcification in vascular smooth muscle cells. Currently, vascular calcification is a silent pathology, and there are no early diagnostic tools. Therefore, by the time vascular calcification is diagnosed, it is usually untreatable. Some mediators, such as oxidative stress, inflammation, and extracellular vesicles, are inducers and promoters of vascular calcification. They play a crucial role during vascular generation and the progression of vascular calcification. Extracellular vesicles, mainly derived from injured endothelial cells that have acquired a senescent phenotype, contribute to calcification in a manner mostly dependent on two factors: (1) the number of extracellular vesicles released, and (2) their cargo. In this review, we present state-of-the-art knowledge on the composition and functions of extracellular vesicles involved in the generation and progression of vascular calcification.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Grupo de Investigación Freshage, Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- Instituto Sanitario de Investigación INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III (CIBERFES, ISCIII), Madrid, Spain
| | - Consuelo Borrás
- Grupo de Investigación Freshage, Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- Instituto Sanitario de Investigación INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III (CIBERFES, ISCIII), Madrid, Spain
- *Correspondence: Consuelo Borrás,
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Matilde Alique,
| |
Collapse
|
11
|
Baicalein and Αlpha-Tocopherol Inhibit Toll-like Receptor Pathways in Cisplatin-Induced Nephrotoxicity. Molecules 2022; 27:molecules27072179. [PMID: 35408581 PMCID: PMC9000769 DOI: 10.3390/molecules27072179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/13/2022] [Accepted: 03/14/2022] [Indexed: 02/05/2023] Open
Abstract
Cisplatin (CP) is a conventional chemotherapeutic agent with serious adverse effects. Its toxicity was linked to the stimulation of oxidative stress and inflammation. As a result, this study explored the protective effect of baicalein and alpha-tocopherol in nephrotoxicity induced by cisplatin. Until receiving an intraperitoneal injection of CP (3 mg/kg BW), rats were given baicalein orally 100 mg/kg for seven days or/and a single intraperitoneal injection of α-tocopherol 250 mg/kg. Renal function was tested to explore whether baicalein and α-tocopherol have any beneficial effects; blood urea nitrogen (BUN), serum creatinine, malondialdehyde (MDA) content, antioxidant activity biomarkers and histopathology of renal tissue, oxidative stress biomarkers, inflammatory response markers, and histopathological features of kidney architecture were measured. Cisplatin treatment resulted in extreme renal failure, as measured by high serum creatinine and BUN levels and severe renal changes. Cisplatin therapy resulted in increased lipid peroxidation and decreased glutathione and superoxide dismutase levels, reflecting oxidative stress. Upon treatment with α-tocopherol, baicalein, and combined therapy, there was augmentation in the antioxidant status as well as a reduction in IL-6, NF-κB, TNF, TLR2, and TLR4 and a significant increase in Keap-1 and NRF-2. The combined treatment was the most effective and the nearest to the normal status. These findings suggest that baicalein and α-tocopherol may be useful in preventing cisplatin-induced nephrotoxicity.
Collapse
|
12
|
Phosphate Burden and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:7-13. [DOI: 10.1007/978-3-030-91623-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Kim JS, Hwang HS. Vascular Calcification in Chronic Kidney Disease: Distinct Features of Pathogenesis and Clinical Implication. Korean Circ J 2021; 51:961-982. [PMID: 34854578 PMCID: PMC8636761 DOI: 10.4070/kcj.2021.0995] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/27/2021] [Accepted: 10/13/2021] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is associated with a higher prevalence of vascular calcification (VC) and cardiovascular disease. VC in CKD patients showed different pathophysiological features from those of the general population. The pathogenesis of VC in CKD is a highly organized process, and prior studies have suggested that patients with CKD have their own specific contributors to the phenotypic change of vascular smooth muscle cells (VSMCs), including uremic toxins, CKD-mineral and bone disease (CKD-MBD), inflammation, and oxidative stress. For the diagnosis and monitoring of VC in CKD, several imaging modalities, including plain radiography, ultrasound, and computed tomography have been utilized. VC in CKD patients has distinct clinical features and implications. CKD patients revealed a more intense and more prevalent calcification on the intimal and medial layers, whereas intimal calcification is predominantly observed in the general population. While a higher VC score is clearly associated with a higher risk of all-cause mortality and cardiovascular events, a greater VC score in CKD patients does not fully reflect the burden of atherosclerosis, because they have more calcification at equal volumes of atheromatous plaques. The primary goal of VC treatment in CKD is the prevention of VC progression, and the main management is to control the biochemical components of CKD-MBD. Cinacalcet and non-calcium-containing phosphate binders are the mainstay of VC prevention in CKD-MBD management. VC in patients with CKD is an ongoing area of research and is expected to advance soon.
Collapse
Affiliation(s)
- Jin Sug Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, Korea
| | - Hyeon Seok Hwang
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
14
|
Inflammation: a putative link between phosphate metabolism and cardiovascular disease. Clin Sci (Lond) 2021; 135:201-227. [PMID: 33416083 PMCID: PMC7796315 DOI: 10.1042/cs20190895] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Dietary habits in the western world lead to increasing phosphate intake. Under physiological conditions, extraosseous precipitation of phosphate with calcium is prevented by a mineral buffering system composed of calcification inhibitors and tight control of serum phosphate levels. The coordinated hormonal regulation of serum phosphate involves fibroblast growth factor 23 (FGF23), αKlotho, parathyroid hormone (PTH) and calcitriol. A severe derangement of phosphate homeostasis is observed in patients with chronic kidney disease (CKD), a patient collective with extremely high risk of cardiovascular morbidity and mortality. Higher phosphate levels in serum have been associated with increased risk for cardiovascular disease (CVD) in CKD patients, but also in the general population. The causal connections between phosphate and CVD are currently incompletely understood. An assumed link between phosphate and cardiovascular risk is the development of medial vascular calcification, a process actively promoted and regulated by a complex mechanistic interplay involving activation of pro-inflammatory signalling. Emerging evidence indicates a link between disturbances in phosphate homeostasis and inflammation. The present review focuses on critical interactions of phosphate homeostasis, inflammation, vascular calcification and CVD. Especially, pro-inflammatory responses mediating hyperphosphatemia-related development of vascular calcification as well as FGF23 as a critical factor in the interplay between inflammation and cardiovascular alterations, beyond its phosphaturic effects, are addressed.
Collapse
|
15
|
Lanthionine, a Novel Uremic Toxin, in the Vascular Calcification of Chronic Kidney Disease: The Role of Proinflammatory Cytokines. Int J Mol Sci 2021; 22:ijms22136875. [PMID: 34206780 PMCID: PMC8269354 DOI: 10.3390/ijms22136875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/24/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Vascular calcification (VC) is a risk factor for cardiovascular events and mortality in chronic kidney disease (CKD). Several components influence the occurrence of VC, among which inflammation. A novel uremic toxin, lanthionine, was shown to increase intracellular calcium in endothelial cells and may have a role in VC. A group of CKD patients was selected and divided into patients with a glomerular filtration rate (GFR) of <45 mL/min/1.73 m2 and ≥45 mL/min/1.73 m2. Total Calcium Score (TCS), based on the Agatston score, was assessed as circulating lanthionine and a panel of different cytokines. A hemodialysis patient group was also considered. Lanthionine was elevated in CKD patients, and levels increased significantly in hemodialysis patients with respect to the two CKD groups; in addition, lanthionine increased along with the increase in TCS, starting from one up to three. Interleukin IL-6, IL-8, and Eotaxin were significantly increased in patients with GFR < 45 mL/min/1.73 m2 with respect to those with GFR ≥ 45 mL/min/1.73 m2. IL-1b, IL-7, IL-8, IL-12, Eotaxin, and VEGF increased in calcified patients with respect to the non-calcified. IL-8 and Eotaxin were elevated both in the low GFR group and in the calcified group. We propose that lanthionine, but also IL-8 and Eotaxin, in particular, are a key feature of VC of CKD, with possible marker significance.
Collapse
|
16
|
Hemodialysis exacerbates proteolytic imbalance and pro-fibrotic platelet dysfunction. Sci Rep 2021; 11:11764. [PMID: 34083719 PMCID: PMC8175411 DOI: 10.1038/s41598-021-91416-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/19/2021] [Indexed: 11/19/2022] Open
Abstract
Multi-organ fibrosis among end stage renal disease (ESRD) patients cannot be explained by uremia alone. Despite mitigation of thrombosis during hemodialysis (HD), subsequent platelet dysfunction and tissue dysregulation are less understood. We comprehensively profiled plasma and platelets from ESRD patients before and after HD to examine HD-modulation of platelets beyond thrombotic activation. Basal plasma levels of proteolytic regulators and fibrotic factors were elevated in ESRD patients compared to healthy controls, with isoform-specific changes during HD. Platelet lysate (PL) RNA transcripts for growth and coagulative factors were elevated post-HD, with upregulation correlated to HD vintage. Platelet secretome correlations to plasma factors reveal acutely induced pro-fibrotic platelet phenotypes in ESRD patients during HD characterized by preferentially enhanced proteolytic enzyme translation and secretion, platelet contribution to inflammatory response, and increasing platelet dysfunction with blood flow rate (BFR) and Vintage. Compensatory mechanisms of increased platelet growth factor synthesis with acute plasma matrix metalloproteinase (MMP) and tissue inhibitor of MMPs (TIMP) increases show short-term mode-switching between dialysis sessions leading to long-term pro-fibrotic bias. Chronic pro-fibrotic adaptation of platelet synthesis were observed through changes in differential secretory kinetics of heterogenous granule subtypes. We conclude that chronic and acute platelet responses to HD contribute to a pro-fibrotic milieu in ESRD.
Collapse
|
17
|
Gupta V, Ekundayo O, Nemeth ZK, Yang Y, Covic A, Mathe Z, Kovesdy CP, Molnar MZ, Mucsi I. Association between serum osteoprotegerin level and mortality in kidney transplant recipients - a prospective observational cohort study. Transpl Int 2021; 34:844-854. [PMID: 33606319 DOI: 10.1111/tri.13847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/07/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Paradoxically, higher serum levels of osteoprotegerin (OPG: a vascular calcification inhibitor) have been associated with increased arterial stiffness, risk of cardiovascular disease and all-cause mortality. A few studies reported that post-transplant OPG levels are associated with mortality in kidney transplant (KT) recipients. In this study, this association was assessed in a cohort of prevalent KT recipients, adjusting for previously untested potential confounders, including fibroblast growth factor 23 (FGF23) and interleukin 6 (IL-6). Socio-demographic and clinical parameters, medical and transplant history, and laboratory data were collected from 982 prevalent KT recipients. The association between serum OPG and all-cause mortality over a 6-year follow-up period was examined using Kaplan-Meier survival curves and multivariable-adjusted Cox regression models. Participants with high serum OPG were more likely female, older, deceased donor KT recipients and have more comorbidity, lower eGFR, higher FGF23, higher IL-6, and longer dialysis vintage. Each 1 pmol/l higher serum OPG level was associated with a 49% higher risk of mortality (hazard ratio (HR) [95% confidence interval (CI)]: 1.49 [1.40-1.61]). This association persisted after adjusting for confounders (HR [95% CI]: 1.20 [1.10-1.30]). In conclusion, serum OPG was associated with all-cause mortality independent of several novel confounders in prevalent KT recipients.
Collapse
Affiliation(s)
- Vardaan Gupta
- Department of Medicine, Division of Nephrology and Multiorgan Transplant Program, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Oladapo Ekundayo
- Department of Medicine, Division of Nephrology and Multiorgan Transplant Program, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Zsofia K Nemeth
- Nephrology Division, Uzsoki Teaching Hospital, Budapest, Hungary
| | - Yifan Yang
- Department of Medicine, Division of Nephrology and Multiorgan Transplant Program, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Adrian Covic
- "C.I. Parhon" University Hospital, Iasi, Romania.,Grigore T, Popa" University of Medicine, Iasi, Romania
| | - Zoltan Mathe
- Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Csaba P Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Miklos Z Molnar
- Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary.,Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Division of Nephrology & Hypertension, Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Istvan Mucsi
- Department of Medicine, Division of Nephrology and Multiorgan Transplant Program, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Chen Z, Zhou Y, Yang T. Histopathological assessment of radial artery calcification in patients with end-stage kidney disease. Ren Fail 2021; 43:362-370. [PMID: 33685341 PMCID: PMC7946001 DOI: 10.1080/0886022x.2021.1889600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background A comprehensive understanding of vascular calcification pathology is significant for the development of cardiovascular disease therapy in high-risk populations. This cross-sectional study aimed to evaluate the prevalence and characteristics of radial artery calcification (RAC) and to identify the factors that are associated with RAC in end-stage kidney disease (ESKD). Methods Detailed medical histories of 180 patients with ESKD were recorded. Fragments of the radial artery obtained during the creation of arteriovenous fistula for hemodialysis access were stained with alizarin red S. Results Calcification was localized in the arterial media layer. The prevalence of positive calcification staining in the radial arteries was 21.1% (n = 38). Patients with RAC had a higher glycated hemoglobin level (p < 0.01), higher prevalence of dialysis duration >5 years (p = 0.022), and diabetes mellitus (p < 0.01) than those without RAC. Multiple logistic regression models showed dialysis duration >5 years (odds ratio [OR], 9.864; 95% confidence interval [CI], 2.666–36.502; p < 0.01) and diabetes mellitus (OR, 12.689; 95% CI, 2.796–34.597; p < 0.01) were independent risk factors for RAC in patients with ESKD. Patients with dialysis duration >5 years had a higher prevalence of RAC (p = 0.012) than those with dialysis duration ≤5 years. Patients with diabetes mellitus had a higher prevalence of RAC (p < 0.01) than those without diabetes mellitus. Patients with diabetes mellitus ≥15 years had a higher prevalence of RAC (p = 0.042) than those with diabetes mellitus <15 years. Radial artery calcification level showed a significantly positive correlation with dialysis duration (p < 0.05), diabetes mellitus duration (p < 0.01), HbA1c level (p < 0.01) and Calcium level (p < 0.01). Conclusions In patients with ESKD, dialysis duration >5 years and diabetes predict RAC. Thus, the combination of prolonged dialysis and hyperglycemic conditions exerts a synergistic effect on RAC.
Collapse
Affiliation(s)
- Zhenwei Chen
- Department of Nephrology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, PR China
| | - Youjian Zhou
- Department of Pathology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, PR China
| | - Tiecheng Yang
- Department of Nephrology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, PR China
| |
Collapse
|
19
|
Nakajima A, Araki M, Kurihara O, Minami Y, Soeda T, Yonetsu T, Higuma T, Kakuta T, McNulty I, Lee H, Malhotra R, Nakamura S, Jang IK. Predictors for Rapid Progression of Coronary Calcification: An Optical Coherence Tomography Study. J Am Heart Assoc 2021; 10:e019235. [PMID: 33496191 PMCID: PMC7955445 DOI: 10.1161/jaha.120.019235] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background The role of coronary calcification in cardiovascular events and plaque stabilization is still being debated, and factors involved in the progression of coronary calcification are not fully understood. This study aimed to identify the predictors for rapid progression of coronary calcification. Methods and Results Patients with serial optical coherence tomography imaging at baseline and at 6 months were selected. Changes in the calcification index and predictors for progression of calcification were studied. Calcification index was defined as the product of the mean calcification arc and calcification length. Rapid progression of calcification was defined as an increase in the calcification index above the median value. Among 187 patients who had serial optical coherence tomography imaging, 235 calcified plaques were identified in 105 patients (56.1%) at baseline. After 6 months, the calcification index increased in 95.3% of calcified plaques from 132.0 to 178.2 (P<0.001). In multivariable analysis, diabetes mellitus (odds ratio [OR], 3.911; P<0.001), chronic kidney disease (OR, 2.432; P=0.037), lipid-rich plaque (OR, 2.698; P=0.034), and macrophages (OR, 6.782; P<0.001) were found to be independent predictors for rapid progression of coronary calcification. Interestingly, rapid progression of calcification was associated with a significant reduction of inflammatory features (thin-cap fibroatheroma; from 21.2% to 11.9%, P=0.003; macrophages; from 74.6% to 61.0%, P=0.001). Conclusions Diabetes mellitus, chronic kidney disease, lipid-rich plaque, and macrophages were independent predictors for rapid progression of coronary calcification. Baseline vascular inflammation and subsequent stabilization may be related to rapid progression of calcification. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT01110538.
Collapse
Affiliation(s)
- Akihiro Nakajima
- Cardiology Division Massachusetts General HospitalHarvard Medical School Boston MA
| | - Makoto Araki
- Cardiology Division Massachusetts General HospitalHarvard Medical School Boston MA
| | - Osamu Kurihara
- Cardiology Division Massachusetts General HospitalHarvard Medical School Boston MA
| | - Yoshiyasu Minami
- Department of Cardiovascular Medicine Kitasato University School of Medicine Sagamihara Kanagawa Japan
| | - Tsunenari Soeda
- Department of Cardiovascular Medicine Nara Medical University Kashihara Nara Japan
| | - Taishi Yonetsu
- Department of Interventional Cardiology Tokyo Medical and Dental University Tokyo Japan
| | - Takumi Higuma
- Division of Cardiology Department of Internal Medicine St. Marianna University School of Medicine Kanagawa Japan
| | - Tsunekazu Kakuta
- Department of Cardiology Tsuchiura Kyodo General Hospital Tsuchiura Ibaraki Japan
| | - Iris McNulty
- Cardiology Division Massachusetts General HospitalHarvard Medical School Boston MA
| | - Hang Lee
- Biostatistics Center Massachusetts General HospitalHarvard Medical School Boston MA
| | - Rajeev Malhotra
- Cardiology Division Massachusetts General HospitalHarvard Medical School Boston MA
| | - Sunao Nakamura
- Interventional Cardiology Unit New Tokyo Hospital Chiba Japan
| | - Ik-Kyung Jang
- Cardiology Division Massachusetts General HospitalHarvard Medical School Boston MA.,Division of Cardiology Kyung Hee University Hospital Seoul Korea
| |
Collapse
|
20
|
Otaki Y, Watanabe T, Konta T, Watanabe M, Asahi K, Yamagata K, Fujimoto S, Tsuruya K, Narita I, Kasahara M, Shibagaki Y, Iseki K, Moriyama T, Kondo M, Watanabe T. Impact of Chronic Kidney Disease on Aortic Disease-related Mortality: A Four-year Community-Based Cohort Study. Intern Med 2021; 60:689-697. [PMID: 33642559 PMCID: PMC7990639 DOI: 10.2169/internalmedicine.5798-20] [Citation(s) in RCA: 3] [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: 12/26/2022] Open
Abstract
Objective Despite advances in medicine, aortic diseases (ADs), such as aneurysm rupture and aortic dissection, remain fatal and carry extremely high mortality rates. Due to its low frequency, the risk of developing AD has not yet been fully elucidated. Chronic kidney disease (CKD) is an established risk factor for cardiovascular disease and mortality. The aim of the present study was to examine whether or not CKD is a risk for AD-related mortality in the general population. Methods We used a nationwide database of 554,442 subjects (40-75 years old) who participated in the annual "Specific Health Check and Guidance in Japan" checkup between 2008 and 2013. Results There were 131 aortic aneurysm and dissection deaths during the follow-up period of 2,123,512 person-years. A Kaplan-Meier analysis revealed that subjects with CKD had a higher rate of AD-related deaths than those without it. A multivariate Cox proportional hazard regression analysis demonstrated that CKD was an independent risk factor for AD-related death in the general population after adjusting for cardiovascular risk factors. The addition of CKD to cardiovascular risk factors significantly improved the C, net reclassification, and integrated discrimination indexes. Conclusion CKD is an additional risk for AD-related death, suggesting that CKD may be a target for the prevention and early identification of subjects at high risk for AD-related death in the general population.
Collapse
Affiliation(s)
- Yoichiro Otaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Tetsu Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Tsuneo Konta
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Masafumi Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Japan
| | - Koichi Asahi
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Kunihiro Yamagata
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Shouichi Fujimoto
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Kazuhiko Tsuruya
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Ichiei Narita
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Masato Kasahara
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Yugo Shibagaki
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Kunitoshi Iseki
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Toshiki Moriyama
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Masahide Kondo
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| | - Tsuyoshi Watanabe
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD) Based on the Individual Risk Assessment by Specific Health Check, Japan
| |
Collapse
|
21
|
Abstract
Phosphorus plays a vital role in diverse biological processes including intracellular signaling, membrane integrity, and skeletal biomineralization; therefore, the regulation of phosphorus homeostasis is essential to the well-being of the organism. Cells and whole organisms respond to changes in inorganic phosphorus (Pi) concentrations in their environment by adjusting Pi uptake and altering biochemical processes in cells (local effects) and distant organs (endocrine effects). Unicellular organisms, such as bacteria and yeast, express specific Pi-binding proteins on the plasma membrane that respond to changes in ambient Pi availability and transduce intracellular signals that regulate the expression of genes involved in cellular Pi uptake. Multicellular organisms, including humans, respond at a cellular level to adapt to changes in extracellular Pi concentrations and also have endocrine pathways which integrate signals from various organs (e.g., intestine, kidneys, parathyroid glands, bone) to regulate serum Pi concentrations and whole-body phosphorus balance. In mammals, alterations in the concentrations of extracellular Pi modulate type III sodium-phosphate cotransporter activity on the plasma membrane, and trigger changes in cellular function. In addition, elevated extracellular Pi induces activation of fibroblast growth factor receptor, Raf/mitogen-activated protein kinase/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) and Akt pathways, which modulate gene expression in various mammalian cell types. Excessive Pi exposure, especially in patients with chronic kidney disease, leads to endothelial dysfunction, accelerated vascular calcification, and impaired insulin secretion.
Collapse
Affiliation(s)
- Kittrawee Kritmetapak
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Division of Nephrology and Hypertension, Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55902, USA
| | - Rajiv Kumar
- Division of Nephrology and Hypertension, Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55902, USA.
- Nephrology Research, Medical Sciences 1-120, 200 First Street Southwest, Rochester, MN, 55902, USA.
| |
Collapse
|
22
|
Yin L, Li X, Ghosh S, Xie C, Chen J, Huang H. Role of gut microbiota-derived metabolites on vascular calcification in CKD. J Cell Mol Med 2020; 25:1332-1341. [PMID: 33369187 PMCID: PMC7875928 DOI: 10.1111/jcmm.16230] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
The interaction between gut microbiota and the host has gained widespread concern. Gut microbiota not only provides nutrients from the ingested food but also generates bioactive metabolites and signalling molecules to impact host physiology, especially in chronic kidney disease (CKD). The development of CKD, accompanied by changed diet and medication, alters the gut flora and causes the effect in distant organs, leading to clinical complications. Vascular calcification (VC) is an actively regulated process and a high prevalence of VC in CKD has also been linked to an imbalance in gut microbiota and altered metabolites. In this review, we focused on gut microbiota-derived metabolites involved in VC in CKD and explained how these metabolites influence the calcification process. Correcting the imbalance of gut microbiota and regulating microbiota-derived metabolites by dietary modification and probiotics are new targets for the improvement of the gut-kidney axis, which indicate innovative treatment options of VC in CKD.
Collapse
Affiliation(s)
- Li Yin
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - XiaoXue Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Sounak Ghosh
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Changming Xie
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jie Chen
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
23
|
Pei J, Ethier I, Hudson RE, Hawley CM, Johnson DW, Campbell SB, Francis RS, Wong G, Craig JC, Viecelli AK, Cho Y. Peritoneal dialysis versus haemodialysis for people commencing dialysis. Hippokratia 2020. [DOI: 10.1002/14651858.cd013800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Juan Pei
- Department of Nephrology; The First Affiliated Hospital of Xiamen University; Xiamen China
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
| | - Isabelle Ethier
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
- Department of Nephrology; Centre hospitalier de l’Université de Montréal; Montréal Canada
| | - Rebecca E Hudson
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
| | - Carmel M Hawley
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
- Australasian Kidney Trials Network; The University of Queensland; Brisbane Australia
- Translational Research Institute; Brisbane Australia
| | - David W Johnson
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
- Australasian Kidney Trials Network; The University of Queensland; Brisbane Australia
- Translational Research Institute; Brisbane Australia
| | - Scott B Campbell
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
| | - Ross S Francis
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
| | - Germaine Wong
- School of Public Health; The University of Sydney; Sydney Australia
| | - Jonathan C Craig
- Cochrane Kidney and Transplant, Centre for Kidney Research; The Children's Hospital at Westmead; Westmead Australia
- College of Medicine and Public Health; Flinders University; Adelaide Australia
| | - Andrea K Viecelli
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
- Australasian Kidney Trials Network; The University of Queensland; Brisbane Australia
- Translational Research Institute; Brisbane Australia
| | - Yeoungjee Cho
- Department of Nephrology; Princess Alexandra Hospital; Brisbane Australia
- Australasian Kidney Trials Network; The University of Queensland; Brisbane Australia
- Translational Research Institute; Brisbane Australia
| |
Collapse
|
24
|
Fasting plasma glucose, lipid ratios, and atherogenic coefficient are the risk factors for hypertension in chronic kidney disease patients on hemodialysis: A report from the Regional High Speciality Hospital of Peninsular Yucatan, Mexico. ANTHROPOLOGICAL REVIEW 2020. [DOI: 10.2478/anre-2020-0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Chronic kidney disease (CKD) is associated with the development of cardivascular disease (CVD). CKD is one of the major public health problems in Mexico. Derived parameters of lipid profile, namely atherogenic index of plasma (AIP), atherogenic coefficient (AC), and Castelli risk index (CRI I and CRI II) are useful for predicting hypertension among CKD patients on hemodialysis that are not widely reported from Mexico. Objective of the present study was to find interrelationships among blood pressure, fasting plasma glucose (FPG), and derived parameters of lipid profile (AIP, AC, CRI-I, and CRI-II) among adult CKD patients on hemodialysis in a hospital in Yucatan, Mexico. Methods: An observational study was performed using the medical records (2016 and 2017) of 47 CKD patients on hemodialysis in the Regional High Speciality Hospital of Yucatan Peninsula (HRAEPY in Spanish acronym). Multiple linear regression models were developed to evaluate the use of FPG level and derived parameters of lipid profile (AC, CRI-I, and CRI-II) as risk factors predicting mean arterial pressure (MAP). Results showed remarkable prevalence of excess weight (55% overweight, 15% obesity) and hypertension (64%) in the sample. Correlation coeffcients and multiple linear regression models showed significant rise of blood pressure in association with elevated FPG level and derived lipid profile parameters. The results confirm the use of FPG, AC, CRI-I and CRI-II as the indicators for an early diagnosis of hypertension and related CVDs among CKD patients on hemodialysis.
Collapse
|
25
|
Elhabashi AF, Sulaibeekh L, Seddiq N, Alali S, Abdulmajeed AK, Perez NS. Presepsin Level Correlates with the Development of Moderate Coronary Artery Calcifications in Hemodialysis Patients: A Preliminary Cross-Section Design Study. Risk Manag Healthc Policy 2020; 13:999-1006. [PMID: 32821182 PMCID: PMC7422906 DOI: 10.2147/rmhp.s262058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/22/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose End-stage renal disease patients have a high mortality rate linked to cardiovascular complications, and one of these complications is vascular calcification. This study was performed to test if presepsin, an inflammatory marker, is a predictor of coronary artery calcification (CAC) in hemodialysis (HD) patients. Patients and Methods This study was a cross-sectional design involving 48 HD patients and 13 control subjects. Coronary artery calcification score (CACs) was evaluated by a high resolution, ECG synchronized computed tomography of the heart using a CT calcium scoring. Presepsin and other laboratory analyses were performed on blood samples drawn before HD. Results Presepsin levels in HD patients were 14 times higher than healthy controls (P<0.01). Also, all laboratory tests except for vitamin D were significantly different than controls. Presepsin, phosphorus levels, and calcium-phosphate product were positively correlated with increasing CACs within groups of zero to moderate calcifications (p<0.05, R=0.459 and <0.01, R=0.591, respectively). These correlations were not seen with eGFR, PTH, calcium, vitamin D, CRP, or ESR levels. Furthermore, the log-transformed data of presepsin correlated with 1–15 months of HD vintage (p<0.05, R=0.482), whereas CACs data correlated with 1–20 months of HD vintage (p<0.05, R=0.425). Conclusion Although this study is preliminary and has a limited number of patients, it shows that presepsin, as an inflammatory marker, correlates with the development of moderate CAC in HD patients and may predict CAC development. Therefore, measuring presepsin and managing inflammation before and during the early phases of HD may lower coronary calcification development. However, more clinical studies in this direction are essential.
Collapse
Affiliation(s)
- Ahmed F Elhabashi
- Bahrain Defense Force Hospital, Royal Medical Services, Riffa, Kingdom of Bahrain
| | - Leena Sulaibeekh
- Bahrain Defense Force Hospital, Royal Medical Services, Riffa, Kingdom of Bahrain
| | - Nahed Seddiq
- Bahrain Defense Force Hospital, Royal Medical Services, Riffa, Kingdom of Bahrain
| | - Salman Alali
- Bahrain Defense Force Hospital, Royal Medical Services, Riffa, Kingdom of Bahrain
| | - Amjad K Abdulmajeed
- Bahrain Defense Force Hospital, Royal Medical Services, Riffa, Kingdom of Bahrain
| | - Nuria S Perez
- Bahrain Defense Force Hospital, Royal Medical Services, Riffa, Kingdom of Bahrain
| |
Collapse
|
26
|
X-ray Micro-Computed Tomography: An Emerging Technology to Analyze Vascular Calcification in Animal Models. Int J Mol Sci 2020; 21:ijms21124538. [PMID: 32630604 PMCID: PMC7352990 DOI: 10.3390/ijms21124538] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification describes the formation of mineralized tissue within the blood vessel wall, and it is highly associated with increased cardiovascular morbidity and mortality in patients with chronic kidney disease, diabetes, and atherosclerosis. In this article, we briefly review different rodent models used to study vascular calcification in vivo, and critically assess the strengths and weaknesses of the current techniques used to analyze and quantify calcification in these models, namely 2-D histology and the o-cresolphthalein assay. In light of this, we examine X-ray micro-computed tomography (µCT) as an emerging complementary tool for the analysis of vascular calcification in animal models. We demonstrate that this non-destructive technique allows us to simultaneously quantify and localize calcification in an intact vessel in 3-D, and we consider recent advances in µCT sample preparation techniques. This review also discusses the potential to combine 3-D µCT analyses with subsequent 2-D histological, immunohistochemical, and proteomic approaches in correlative microscopy workflows to obtain rich, multifaceted information on calcification volume, calcification load, and signaling mechanisms from within the same arterial segment. In conclusion we briefly discuss the potential use of µCT to visualize and measure vascular calcification in vivo in real-time.
Collapse
|
27
|
Torino C, Gargani L, Sicari R, Letachowicz K, Ekart R, Fliser D, Covic A, Siamopoulos K, Stavroulopoulos A, Massy ZA, Fiaccadori E, Regolisti G, Bachelet T, Slotki I, Martinez-Castelao A, Coudert-Krier MJ, Rossignol P, Hannedouche T, Wiecek A, Sarafidis P, Battaglia Y, Prohić N, Klinger M, Hojs R, Seiler-Mußler S, Lizzi F, Siriopol D, Balafa O, Shavit L, Loutradis C, Seidowsky A, Tripepi R, Mallamaci F, Tripepi G, Picano E, London GM, Zoccali C. Inflammation is an amplifier of lung congestion by high lv filling pressure in hemodialysis patients: a longitudinal study. J Nephrol 2020; 33:583-590. [PMID: 31916229 DOI: 10.1007/s40620-019-00696-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/26/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Since inflammation alters vascular permeability, including vascular permeability in the lung, we hypothesized that it can be an amplifier of lung congestion in a category of patients at high risk for pulmonary oedema like end stage kidney disease (ESKD) patients. OBJECTIVE AND METHODS We investigated the effect modification by systemic inflammation (serum CRP) on the relationship between a surrogate of the filling pressure of the LV [left atrial volume indexed to the body surface area (LAVI)] and lung water in a series of 220 ESKD patients. Lung water was quantified by the number of ultrasound B lines (US-B) on lung US. Six-hundred and three recordings were performed during a 2-year follow up. Longitudinal data analysis was made by the Mixed Linear Model. RESULTS At baseline, 88 had absent, 101 had mild to moderate lung congestion and 31 severe congestion. The number of US B lines associated with LAVI (r = 0.23, P < 0.001) and serum CRP was a robust modifier of this relationship (P < 0.001). Similarly, in fully adjusted longitudinal analyses US-B lines associated with simultaneous estimates of LAVI (P = 0.002) and again CRP was a strong modifier of this relationship in adjusted analyses (P ≤ 0.01). Overall, at comparable LAVI levels, lung congestion was more pronounced in inflamed than in non-inflamed patients. CONCLUSION In ESKD systemic inflammation is a modifier of the relationship between LAVI, an integrate measure of LV filling pressure, and lung water. For any given pressure, lung water is increased with higher CRP levels, likely reflecting a higher permeability of the alveolar-capillary barrier.
Collapse
Affiliation(s)
- Claudia Torino
- Institute of Clinical Physiology-Reggio Cal Unit, National Research Council, Reggio Calabria, Italy
| | - Luna Gargani
- Institute of Clinical Physiology-Pisa, National Research Council, Pisa, Italy
| | - Rosa Sicari
- Institute of Clinical Physiology-Pisa, National Research Council, Pisa, Italy
| | | | - Robert Ekart
- University Clinical Centre Maribor, Maribor, Slovenia
| | | | | | | | | | - Ziad A Massy
- Ambroise Pare University Hospital, APHP, Paris-Ile-de France-Ouest University (UVSQ), INSERM U1018, Boulogne Billancourt, Paris, France
| | - Enrico Fiaccadori
- Dipartiment of Medicine and Surgery and Nephrology Unit, University Hospital Parma, Parma, Italy
| | - Giuseppe Regolisti
- Dipartiment of Medicine and Surgery and Nephrology Unit, University Hospital Parma, Parma, Italy
| | | | | | - Alberto Martinez-Castelao
- Bellvitge's University Hospital-Hospitalet, Barcelona, REDinREN Instituto Salud Carlos III, Madrid, Spain
| | - Marie-Jeanne Coudert-Krier
- Centre D'Investigation Clinique Plurithématique Pierre Drouin-INSERM-CHRU de Nancy-Université de Lorraine and Institute Lorrain du Coeur Et de Vaisseaux Louis Mathieu, Vandœuvre-lès-Nancy, France
| | - Patrick Rossignol
- Centre D'Investigation Clinique Plurithématique Pierre Drouin-INSERM-CHRU de Nancy-Université de Lorraine and Institute Lorrain du Coeur Et de Vaisseaux Louis Mathieu, Vandœuvre-lès-Nancy, France
- FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists) Network, Nancy, France
| | - Thierry Hannedouche
- University Hospital Strasbourg, Strasbourg, France
- FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists) Network, Nancy, France
| | | | | | | | - Nejra Prohić
- Clinical Centre University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Radovan Hojs
- University Clinical Centre Maribor, Maribor, Slovenia
| | | | - Fabio Lizzi
- Saarland University Hospital, Homburg, Germany
| | | | - Olga Balafa
- University Hospital of Ioannina, Ioannina, Greece
| | | | | | - Alexandre Seidowsky
- Ambroise Pare University Hospital, APHP, Paris-Ile-de France-Ouest University (UVSQ), INSERM U1018, Boulogne Billancourt, Paris, France
| | - Rocco Tripepi
- Institute of Clinical Physiology-Reggio Cal Unit, National Research Council, Reggio Calabria, Italy
| | - Francesca Mallamaci
- Institute of Clinical Physiology-Reggio Cal Unit, National Research Council, Reggio Calabria, Italy
| | - Giovanni Tripepi
- Institute of Clinical Physiology-Reggio Cal Unit, National Research Council, Reggio Calabria, Italy
| | - Eugenio Picano
- Institute of Clinical Physiology-Pisa, National Research Council, Pisa, Italy
| | - Gérard Michel London
- Centre Hospitalier F.H. Manhès, Fleury-Mérogis, France
- FCRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists) Network, Nancy, France
| | - Carmine Zoccali
- Institute of Clinical Physiology-Reggio Cal Unit, National Research Council, Reggio Calabria, Italy.
- CNR-IFC, Clinical Epidemiology of Renal Diseases and Hypertension, 89124, Reggio Calabria, Italy.
| |
Collapse
|
28
|
Pang H, Xiao L, Lu Z, Chen H, Shang Z, Jiang N, Wang X, Wei F, Jiang A, Chen Y, Niu Y. Targeting androgen receptor in macrophages inhibits phosphate-induced vascular smooth muscle cell calcification by decreasing IL-6 expression. Vascul Pharmacol 2020; 130:106681. [PMID: 32387336 DOI: 10.1016/j.vph.2020.106681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/24/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is a common complication of chronic kidney disease (CKD). However, its mechanisms remain unclear. VC, similar to atherosclerosis, is an inflammatory disease. Vascular smooth muscle cells (VSMCs) play a key role in VC progression. The androgen receptor (AR) in monocytes/macrophages plays an important role in inflammatory diseases. Here, we define the role of macrophage (MФ) AR in inorganic phosphate-induced VSMC calcification. Our results show that the conditioning medium (CM) of silencing AR in macrophages inhibits inorganic phosphate-induced human aortic smooth muscle cell (HASMC) calcification, and alleviates the transdifferentiation of HASMCs into osteoblasts for the protein expression of osteoblasts marker Runt-related transcription factor-2 (Runx2) in HASMCs decreased while that of smooth muscle cell marker SM22α increased. The effect of AR on HASMC calcification might mainly be mediated by the inflammatory cytokine IL-6. Silencing AR in monocytes/macrophages can dramatically decrease IL-6 expression. We also investigated how macrophage AR regulates IL-6. ChIP and luciferase assays indicate that AR directly binds to the ARE sequence in the promoter of the IL-6 gene to accelerate transcription and expression. To our knowledge, this is the first investigation that has established the correlation between AR and VC and identified the contribution of AR in the calcification of VSMCs. In addition, this study describes a novel target for therapeutic intervention in VC.
Collapse
Affiliation(s)
- Haiyan Pang
- The Kidney Diseases and Blood Purification Center, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Longfei Xiao
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Zhi Lu
- The Kidney Diseases and Blood Purification Center, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Haiyan Chen
- The Kidney Diseases and Blood Purification Center, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Zhiqun Shang
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Ning Jiang
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Xiaojuan Wang
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Fang Wei
- The Kidney Diseases and Blood Purification Center, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Aili Jiang
- The Kidney Diseases and Blood Purification Center, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China
| | - Yegang Chen
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China.
| | - Yuanjie Niu
- Department of Urology, Tianjin Institute of Urology, Tianjin Medical University Second Hospital, Tianjin 300211, China.
| |
Collapse
|
29
|
An overview of the mechanisms in vascular calcification during chronic kidney disease. Curr Opin Nephrol Hypertens 2020; 28:289-296. [PMID: 30985336 DOI: 10.1097/mnh.0000000000000507] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Chronic kidney disease (CKD) facilitates a unique environment to strongly accelerate vascular calcification - the pathological deposition of calcium-phosphate in the vasculature. These calcifications are associated with the excessive cardiovascular mortality of CKD patients. RECENT FINDINGS Vascular calcification is a multifaceted active process, mediated, at least partly, by vascular smooth muscle cells. These cells are able to transdifferentiate into cells with osteo/chondrogenic properties, which exert multiple effects to facilitate vascular tissue mineralization. As the understanding of the underlying pathophysiology increases, first therapeutic concepts begin to emerge. SUMMARY This brief review provides an overview on the so far known mechanisms involved in the initiation and progression of vascular calcification in CKD.
Collapse
|
30
|
Timofte D, Dragoș D, Balcangiu-Stroescu AE, Tănăsescu MD, Gabriela Bălan D, Răducu L, Tulin A, Stiru O, Ionescu D. Abdominal aortic calcification in predialysis patients: Contribution of traditional and uremia-related risk factors. Exp Ther Med 2020; 20:97-102. [PMID: 32509000 PMCID: PMC7271689 DOI: 10.3892/etm.2020.8607] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease is the main cause of morbidity and mortality in end-stage renal disease (ESRD) patients. Recent population based epidemiological studies demonstrated a correlation between reduced renal function and risk for all causes and cardiovascular mortality, 50% of patients dying before the commencement of renal replacement therapy. Vascular calcifications (VC) and arterial stiffness are major contributors to cardiovascular disease and are independent predictors of cardiovascular mortality in ESRD patients. Scarce information is available on the risk factors and prognosis of predialysis patients with VC. The aim of this study was to evaluate the contribution of traditional and uremia related risk factors to abdominal aortic calcification in predialysis patients. A single center, retrospective study was performed on 305 adult patients monitored at the Bucharest University Emergency Hospital for at least 6 months. Our study reports an increased incidence of VC in predialysis chronic kidney disease (CKD) patients, higher in those with advanced age, history of cardiovascular disease, osteoporosis, and declining renal function.
Collapse
Affiliation(s)
- Delia Timofte
- Department of Dialysis, Emergency University Hospital, 050098 Bucharest, Romania
| | - Dorin Dragoș
- Department of Medical Semiology, Discipline of Internal Medicine I and Nephrology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Nephrology, Emergency University Hospital, 050098 Bucharest, Romania
| | - Andra-Elena Balcangiu-Stroescu
- Department of Dialysis, Emergency University Hospital, 050098 Bucharest, Romania.,Discipline of Physiology, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Maria-Daniela Tănăsescu
- Department of Medical Semiology, Discipline of Internal Medicine I and Nephrology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Nephrology, Emergency University Hospital, 050098 Bucharest, Romania
| | - Daniela Gabriela Bălan
- Discipline of Physiology, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Laura Răducu
- Department of Plastic and Reconstructive Microsurgery, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Plastic and Reconstructive Surgery, 'Prof. Dr. Agrippa Ionescu' Clinical Emergency Hospital, 011356 Bucharest, Romania
| | - Adrian Tulin
- Department of Anatomy, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of General Surgery, 'Prof. Dr. Agrippa Ionescu' Clinical Emergency Hospital, 011356 Bucharest, Romania
| | - Ovidiu Stiru
- Department of Cardiovascular Surgery, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Cardiovascular Surgery, 'Prof. Dr. C.C. Iliescu' Emergency Institute for Cardiovascular Diseases, 022322 Bucharest, Romania
| | - Dorin Ionescu
- Department of Medical Semiology, Discipline of Internal Medicine I and Nephrology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Nephrology, Emergency University Hospital, 050098 Bucharest, Romania
| |
Collapse
|
31
|
Zhang X, Li Y, Yang P, Liu X, Lu L, Chen Y, Zhong X, Li Z, Liu H, Ou C, Yan J, Chen M. Trimethylamine-N-Oxide Promotes Vascular Calcification Through Activation of NLRP3 (Nucleotide-Binding Domain, Leucine-Rich-Containing Family, Pyrin Domain-Containing-3) Inflammasome and NF-κB (Nuclear Factor κB) Signals. Arterioscler Thromb Vasc Biol 2020; 40:751-765. [DOI: 10.1161/atvbaha.119.313414] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Objectives:
Vascular calcification is highly prevalent in patients with chronic kidney disease. Increased plasma trimethylamine N-oxide (TMAO), a gut microbiota-dependent product, concentrations are found in patients undergoing hemodialysis. However, a clear mechanistic link between TMAO and vascular calcification is not yet established. In this study, we investigate whether TMAO participates in the progression of vascular calcification using in vitro, ex vivo, and in vivo models.
Approach and Results:
Alizarin red staining revealed that TMAO promoted calcium/phosphate-induced calcification of rat and human vascular smooth muscle cells in a dose-dependent manner, and this was confirmed by calcium content assay. Similarly, TMAO upregulated the expression of bone-related molecules including Runx2 (Runt-related transcription factor 2) and BMP2 (bone morphogenetic protein-2), suggesting that TMAO promoted osteogenic differentiation of vascular smooth muscle cells. In addition, ex vivo study also showed the positive regulatory effect of TMAO on vascular calcification. Furthermore, we found that TMAO accelerated vascular calcification in rats with chronic kidney disease, as indicated by Mico-computed tomography analysis, alizarin red staining and calcium content assay. By contrast, reducing TMAO levels by antibiotics attenuated vascular calcification in chronic kidney disease rats. Interestingly, TMAO activated NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome and NF-κB (nuclear factor κB) signals during vascular calcification. Inhibition of NLRP3 inflammasome and NF-κB signals attenuated TMAO-induced vascular smooth muscle cell calcification.
Conclusions:
This study for the first time demonstrates that TMAO promotes vascular calcification through activation of NLRP3 inflammasome and NF-κB signals, suggesting the potential link between gut microbial metabolism and vascular calcification. Reducing the levels of TMAO could become a potential treatment strategy for vascular calcification in chronic kidney disease.
Collapse
Affiliation(s)
- Xiuli Zhang
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Yining Li
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Pingzhen Yang
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Xiaoyu Liu
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Lihe Lu
- Department of Pathophysiolgy, Zhongshan Medical School, Sun Yat-Sen University, China (L.L., Y.C.)
| | - Yanting Chen
- Department of Pathophysiolgy, Zhongshan Medical School, Sun Yat-Sen University, China (L.L., Y.C.)
| | - Xinglong Zhong
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Zehua Li
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Hailin Liu
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Caiwen Ou
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Jianyun Yan
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| | - Minsheng Chen
- From the Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
- Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, China (X.Z., Y.L., P.Y., X.L., X.Z., Z.L., H.L., C.O., J.Y., M.C.)
| |
Collapse
|
32
|
Shioi A, Morioka T, Shoji T, Emoto M. The Inhibitory Roles of Vitamin K in Progression of Vascular Calcification. Nutrients 2020; 12:nu12020583. [PMID: 32102248 PMCID: PMC7071387 DOI: 10.3390/nu12020583] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Vitamin K is a fat-soluble vitamin that is indispensable for the activation of vitamin K-dependent proteins (VKDPs) and may be implicated in cardiovascular disease (CVD). Vascular calcification is intimately associated with CV events and mortality and is a chronic inflammatory process in which activated macrophages promote osteoblastic differentiation of vascular smooth muscle cells (VSMCs) through the production of proinflammatory cytokines such as IL-1β, IL-6, TNF-α, and oncostatin M (OSM) in both intimal and medial layers of arterial walls. This process may be mainly mediated through NF-κB signaling pathway. Vitamin K has been demonstrated to exert anti-inflammatory effects through antagonizing NF-κB signaling in both in vitro and in vivo studies, suggesting that vitamin K may prevent vascular calcification via anti-inflammatory mechanisms. Matrix Gla protein (MGP) is a major inhibitor of soft tissue calcification and contributes to preventing both intimal and medial vascular calcification. Vitamin K may also inhibit progression of vascular calcification by enhancing the activity of MGP through facilitating its γ-carboxylation. In support of this hypothesis, the procalcific effects of warfarin, an antagonist of vitamin K, on arterial calcification have been demonstrated in several clinical studies. Among the inactive MGP forms, dephospho-uncarboxylated MGP (dp-ucMGP) may be regarded as the most useful biomarker of not only vitamin K deficiency, but also vascular calcification and CVD. There have been several studies showing the association of circulating levels of dp-ucMGP with vitamin K intake, vascular calcification, mortality, and CVD. However, additional larger prospective studies including randomized controlled trials are necessary to confirm the beneficial effects of vitamin K supplementation on CV health.
Collapse
Affiliation(s)
- Atsushi Shioi
- Department of Vascular Medicine and Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan;
- Correspondence: ; Tel.: +81666453931
| | - Tomoaki Morioka
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka 545-85858, Japan; (T.M.); (M.E.)
| | - Tetsuo Shoji
- Department of Vascular Medicine and Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan;
| | - Masanori Emoto
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka 545-85858, Japan; (T.M.); (M.E.)
| |
Collapse
|
33
|
Liao R, Wang L, Li J, Sun S, Xiong Y, Li Y, Han M, Jiang H, Anil M, Su B. Vascular calcification is associated with Wnt-signaling pathway and blood pressure variability in chronic kidney disease rats. Nephrology (Carlton) 2019; 25:264-272. [PMID: 31709686 DOI: 10.1111/nep.13677] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/29/2019] [Accepted: 10/27/2019] [Indexed: 02/05/2023]
Abstract
AIM Vascular calcification (VC) is a common complication in chronic kidney disease (CKD) and has been shown to be associated with increased cardiovascular events and mortality. This study was to explore the role of Wnt-signaling pathway in CKD VC, and the association between VC and blood pressure variability (BPV) which is a risk factor of cardiovascular events. METHODS Adult male Sprague-Dawley rats were divided into adenine-induced CKD group (n = 5), 5/6 nephrectomy CKD group (n = 5), sham group (n = 5) and control group (n = 5). Low-calcium-high-phosphate diets were introduced to induce vascular calcification. Both daytime (hour-to-hour during the day) and mid-term (day-to-day for 9 days) blood pressure (BP) were collected and analyzed for BPV metrics. At sacrifice, kidney, heart and aorta samples were taken for histological analyses. Calcium deposition in aorta was identified with Alizarin Red stain and graded. Immunohistochemistry stain and western blot were performed for Wnt3a, Wnt5a, β-catenin, sclerostin, osteopontin, and α-SMA. RESULTS Compared with control rats, CKD rats suffered from markedly severer VC (Grade 2.6 ± 0.2 and 1.8 ± 0.8 vs 0.0 ± 0.0 and 0.2 ± 0.4, P = .0010). VC was positively correlated with vascular Wnt3a and β-catenin expression (P = .0032 and .0000), but not significantly associated with Wnta5a or sclerostin. Besides, CKD rats showed increased BPV (P < .001), which was also positively correlated with VC. CONCLUSION We confirmed that CKD rats had enhanced Wnt-signaling in vascular tissue and severer aorta calcification together with increased BPV. Wnt pathway may be a potential target in future VC and BPV management in CKD.
Collapse
Affiliation(s)
- Ruoxi Liao
- National Clinical Research Center for Geriatrics and Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China.,Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Liya Wang
- National Clinical Research Center for Geriatrics and Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China.,Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiameng Li
- National Clinical Research Center for Geriatrics and Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China.,Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Si Sun
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqin Xiong
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Yupei Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Mei Han
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Heng Jiang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Mahajan Anil
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Rodríguez-Carrio J, Carrillo-López N, Ulloa C, Seijo M, Rodríguez-García M, Rodríguez-Suárez C, Díaz-Corte C, Cannata-Andía JB, Suárez A, Dusso AS. A subset of low density granulocytes is associated with vascular calcification in chronic kidney disease patients. Sci Rep 2019; 9:13230. [PMID: 31519925 PMCID: PMC6744494 DOI: 10.1038/s41598-019-49429-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/24/2019] [Indexed: 01/11/2023] Open
Abstract
Inflammation is central to chronic kidney disease (CKD) pathogenesis and vascular outcomes, but the exact players remain unidentified. Since low density granulocytes (LDGs) are emerging mediators in inflammatory conditions, we aimed to evaluate whether LDGs may be altered in CKD and related to clinical outcomes as biomarkers. To his end, LDGs subsets were measured in peripheral blood by flow cytometry and confocal microscopy in 33 CKD patients undergoing peritoneal dialysis and 15 healthy controls (HC). Analyses were replicated in an additional cohort. DEF3 (marker of early granulopoiesis) gene expression on PBMCs was quantified by qPCR. Total CD15+ LDGs and both CD14lowCD16+ and CD14-CD16- subsets were expanded in CKD. The relative frequency of the CD14-CD16- subpopulation was higher among the CD15+ pool in CKD. This alteration was stable over-time. The increased CD14-CD16-CD15+ paralleled Kauppila scores and DEF3 expression, whereas no association was found with CD14lowCD16+ CD15+. Both subsets differed in their CD11b, CD10, CD35, CD31, CD62L, IFNAR1 and CD68 expression, FSC/SSC features and nuclear morphology, pointing to different origins and maturation status. In conclusion, LDGs were expanded in CKD showing a skewed distribution towards a CD14-CD16-CD15+ enrichment, in association with vascular calcification. DEF3 expression in PBMC can be a marker of LDG expansion.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Catalina Ulloa
- Division of Nephrology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Mariana Seijo
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Laboratorio de Enfermedades Metabólicas Óseas, Hospital de Clínicas, Instituto de Inmunología, Genética y Metabolismo (INIGEM) CONICET- UBA, Buenos Aires, Argentina
| | - Minerva Rodríguez-García
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Spain
- Division of Nephrology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Carmen Díaz-Corte
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Spain
- Division of Nephrology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Spain
- Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Ana Suárez
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Spain.
| | - Adriana S Dusso
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III (ISCIII), Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
35
|
Determinants and Prevention of Coronary Disease in Patients With Chronic Kidney Disease. Can J Cardiol 2019; 35:1181-1187. [DOI: 10.1016/j.cjca.2019.05.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/09/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
|
36
|
Ma K, Liu P, Al-Maghout T, Sukkar B, Cao H, Voelkl J, Alesutan I, Pieske B, Lang F. Phosphate-induced ORAI1 expression and store-operated Ca 2+ entry in aortic smooth muscle cells. J Mol Med (Berl) 2019; 97:1465-1475. [PMID: 31385016 DOI: 10.1007/s00109-019-01824-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 07/02/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022]
Abstract
Compromised renal phosphate elimination in chronic kidney disease (CKD) leads to hyperphosphatemia, which in turn triggers osteo-/chondrogenic signaling in vascular smooth muscle cells (VSMCs) and vascular calcification. Osteo-/chondrogenic transdifferentiation of VSMCs leads to upregulation of the transcription factors MSX2, CBFA1, and SOX9 as well as tissue-nonspecific alkaline phosphatase (ALPL) which fosters calcification by degrading the calcification inhibitor pyrophosphate. Osteo-/chondrogenic signaling in VSMCs involves the serum- and glucocorticoid-inducible kinase SGK1. As shown in other cell types, SGK1 is a powerful stimulator of ORAI1, a Ca2+-channel accomplishing store-operated Ca2+-entry (SOCE). ORAI1 is stimulated following intracellular store depletion by the Ca2+ sensor STIM1. The present study explored whether phosphate regulates ORAI1 and/or STIM1 expression and, thus, SOCE in VSMCs. To this end, primary human aortic smooth muscle cells (HAoSMCs) were exposed to the phosphate donor β-glycerophosphate. Transcript levels were estimated by qRT-PCR, protein abundance by western blotting, ALPL activity by colorimetry, calcification by alizarin red S staining, cytosolic Ca2+-concentration ([Ca2+]i) by Fura-2-fluorescence, and SOCE from increase of [Ca2+]i following re-addition of extracellular Ca2+ after store depletion with thapsigargin. As a result, β-glycerophosphate treatment increased ORAI1 and STIM1 transcript levels and protein abundance as well as SOCE in HAoSMCs. Additional treatment with ORAI1 inhibitor MRS1845 or SGK1 inhibitor GSK650394 virtually disrupted the effects of β-glycerophosphate on SOCE. Moreover, the β-glycerophosphate-induced MSX2, CBFA1, SOX9, and ALPL mRNA expression and activity in HAoSMCs were suppressed in the presence of the ORAI1 inhibitor and upon ORAI1 silencing. In conclusion, enhanced phosphate upregulates ORAI1 and STIM1 expression and store-operated Ca2+-entry, which participate in the orchestration of osteo-/chondrogenic signaling of VSMCs. KEY MESSAGES: • In aortic SMC, phosphate donor ß-glycerophosphate upregulates Ca2+ channel ORAI1. • In aortic SMC, ß-glycerophosphate upregulates ORAI1-activator STIM1. • In aortic SMC, ß-glycerophosphate upregulates store-operated Ca2+-entry (SOCE). • The effect of ß-glycerophosphate on SOCE is disrupted by ORAI1 inhibitor MRS1845. • Stimulation of osteogenic signaling is disrupted by MRS1845 and ORAI1 silencing.
Collapse
Affiliation(s)
- Ke Ma
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Ping Liu
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Tamer Al-Maghout
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Basma Sukkar
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Hang Cao
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040, Linz, Austria.,Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Department of Nephrology and Medical Intensive Care, Charité University Medicine, Berlin, Germany
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040, Linz, Austria.,Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Florian Lang
- Department of Vegetative and Clinical Physiology, University of Tübingen, Wilhelmstr. 56, 72074, Tübingen, Germany.
| |
Collapse
|
37
|
The Emerging Role of Mesenchymal Stem Cells in Vascular Calcification. Stem Cells Int 2019; 2019:2875189. [PMID: 31065272 PMCID: PMC6466855 DOI: 10.1155/2019/2875189] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/12/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
Vascular calcification (VC), characterized by hydroxyapatite crystal depositing in the vessel wall, is a common pathological condition shared by many chronic diseases and an independent risk factor for cardiovascular events. Recently, VC is regarded as an active, dynamic cell-mediated process, during which calcifying cell transition is critical. Mesenchymal stem cells (MSCs), with a multidirectional differentiation ability and great potential for clinical application, play a duplex role in the VC process. MSCs facilitate VC mainly through osteogenic transformation and apoptosis. Meanwhile, several studies have reported the protective role of MSCs. Anti-inflammation, blockade of the BMP2 signal, downregulation of the Wnt signal, and antiapoptosis through paracrine signaling are possible mechanisms. This review displays the evidence both on the facilitating role and on the protective role of MSCs, then discusses the key factors determining this divergence.
Collapse
|
38
|
A dual role for GRP in cardiovascular disease. Aging (Albany NY) 2019; 11:1323-1324. [PMID: 30852562 PMCID: PMC6428097 DOI: 10.18632/aging.101851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/05/2019] [Indexed: 01/07/2023]
|