1
|
Kara AV, Inan H, Durmaz O, Ozdemir R. The relationship between serum uric acid level and carotid intima-media thickness in hemodialysis patients. Hemodial Int 2024; 28:397-404. [PMID: 39098818 DOI: 10.1111/hdi.13174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/06/2024]
Abstract
INTRODUCTION We aimed to evaluate the relationship between carotid intima-media thickness (CIMT), which is a known indicator of cardiovascular risk and atherosclerosis, and uric acid level, which may be an easy marker for cardiovascular diseases due to its antioxidant and pro-oxidant properties in hemodialysis patients. METHODS In this cross-sectional study, we evaluated 77 hemodialysis patients. The mean CIMT of these patients was measured and recorded by Doppler ultrasonography. Patients were divided into two groups according to their serum uric acid levels. Correlation analysis and linear regression analysis were used to define the relationship between study parameters. FINDINGS The mean CIMT levels in the normouricemic group and the hyperuricemic group were 0.95 ± 0.15 and 1.07 ± 0.15, respectively. There was a statistically significant difference between the two groups (p = 0.001). There was a statistically significant and moderate linear correlation between serum uric acid level and mean CIMT (r = 0.402; p = 0.002). Univariate and multivariate linear regression analyses were performed to identify variables that could independently affect the mean CIMT value. According to analysis, uric acid (p < 0.001), hypertension (p = 0.008), albumin (p = 0.029), and C-reactive protein (p = 0.042) were found independent risk factors for mean CIMT value. DISCUSSION We found a significant relationship between serum uric acid level and CIMT, which indicates carotid atherosclerosis. Serum uric acid level is a low-cost laboratory parameter that can be measured in almost all laboratories, and it may be valuable in the hemodialysis patient group to identify patients at high risk of carotid atherosclerosis.
Collapse
Affiliation(s)
- Ali Veysel Kara
- Faculty of Medicine, Department of Nephrology, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Hamza Inan
- Faculty of Medicine, Department of Internal Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Onder Durmaz
- Faculty of Medicine, Department of Radiology, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Ridvan Ozdemir
- Faculty of Medicine, Department of Nephrology, Erzincan Binali Yildirim University, Erzincan, Turkey
| |
Collapse
|
2
|
Tang H, Chen H, Li Z, Xu S, Yan G, Tang C, Liu H. Association between uric acid level and contrast-induced acute kidney injury in patients with type 2 diabetes mellitus after coronary angiography: a retrospective cohort study. BMC Nephrol 2022; 23:399. [PMID: 36510177 PMCID: PMC9746209 DOI: 10.1186/s12882-022-03030-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND This study assessed the predictive value of uric acid (UA) for contrast-induced acute kidney injury (CI-AKI) in patients with type 2 diabetes mellitus (T2DM) who underwent coronary angiography (CAG). A nomogram to aid in the prediction of CI-AKI was also developed and validated, and the construction of a prognostic nomogram combined with clinical features was attempted. METHODS This study retrospectively enrolled T2DM patients who underwent CAG between December 2019 and December 2020 at the Affiliated Zhongda Hospital of Southeast University. Multivariable logistic regression analysis was used for the analysis of clinical outcomes. Receiver operating characteristic (ROC) analyses were performed to determine the area under the ROC curve (AUC) and the cut-off points for continuous clinical data. The prediction accuracies of models for CI-AKI were estimated through Harrell's concordance indices (C-index). Nomograms of the prognostic models were plotted for individualized evaluations of CI-AKI in T2DM patients after CAG. RESULTS A total of 542 patients with T2DM who underwent CAG were included in this study. We found that a high UA level (≥ 425.5 µmol/L; OR = 6.303), BUN level (≥ 5.98 mmol/L; OR = 3.633), Scr level (≥ 88.5 µmol/L; OR = 2.926) and HbA1C level (≥ 7.05%; OR = 5.509) were independent factors for CI-AKI in T2DM patients after CAG. The nomogram model based on UA, BUN, Scr and HbA1C levels presented outstanding performance for CI-AKI prediction (C-index: 0.878). Decision curve analysis (DCA) showed good clinical applicability in predicting the incidence of CI-AKI in T2DM patients who underwent CAG. CONCLUSION High UA levels are associated with an increased incidence of CI-AKI in T2DM patients after CAG. The developed nomogram model has potential predictive value for CI-AKI and might serve as an economic and efficient prognostic tool in clinical practice.
Collapse
Affiliation(s)
- Haixia Tang
- grid.263826.b0000 0004 1761 0489Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu China
| | - Haoying Chen
- grid.452858.60000 0005 0368 2155Department of Ultrasonography, Taizhou central hospital, Taizhou university hospital, Ningbo, China
| | - Zuolin Li
- grid.263826.b0000 0004 1761 0489Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu China
| | - Shengchun Xu
- grid.263826.b0000 0004 1761 0489Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu China
| | - Gaoliang Yan
- grid.263826.b0000 0004 1761 0489Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu China
| | - Chengchun Tang
- grid.263826.b0000 0004 1761 0489Department of Cardiology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu China
| | - Hong Liu
- grid.263826.b0000 0004 1761 0489Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu China
| |
Collapse
|
3
|
Protective Effects of Astragaloside IV on Uric Acid-Induced Pancreatic β-Cell Injury through PI3K/AKT Pathway Activation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2429162. [PMID: 35047042 PMCID: PMC8763508 DOI: 10.1155/2022/2429162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/30/2021] [Accepted: 12/24/2021] [Indexed: 11/25/2022]
Abstract
Background Elevated uric acid (UA) has been found to damage pancreatic β-cell, promote oxidative stress, and cause insulin resistance in type 2 diabetes (T2D). Astragaloside IV (AS-IV), a major active monomer extracted from Astragalus membranaceus (Fisch.) Bunge. which belongs to TRIB. Galegeae (Br.) Torrey et Gray, Papilionaceae, exhibits various activities in a pathophysiological environment and has been widely employed to treat diseases. However, the effects of AS-IV on UA-induced pancreatic β-cell damage need to be investigated and the associating mechanism needs to be elucidated. This study was designed to determine the protective effects and underlying mechanism of AS-IV on UA-induced pancreatic β-cell dysfunction in T2D. Methods UA-treated Min6 cells were exposed to AS-IV or wortmannin. Thereafter, the 3-(45)-dimethylthiahiazo(-z-y1)-35-di-phenytetrazoliumromide (MTT) assay and flow cytometry were employed to determine the effect of AS-IV on cell proliferation and apoptosis, respectively. Insulin secretion was evaluated using the glucose-stimulated insulin secretion (GSIS) assay. Finally, western blot and quantitative real-time polymerase chain reaction (qRT-PCR) were performed to determine the effect of AS-IV on the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway in UA-treated cells. Results AS-IV had no cytotoxic effects on Min6 cells. UA significantly suppressed Min6 cell growth, promoted cell apoptosis, and enhanced caspase-3 activity; however, AS-IV abolished these effects in a dose-dependent manner. Further, decreased insulin secretion was found in UA-treated Min6 cells compared to control cells, and the production of insulin was enhanced by AS-IV in a dose-dependent manner. AS-IV significantly increased phosphorylated (p)-AKT expression and the ratio of p-AKT/AKT in Min6 cells exposed to UA. No evident change in AKT mRNA level was found in the different groups. However, the effects of AS-IV on UA-stimulated Min6 cells were reversed by 100 nM wortmannin. Conclusion Collectively, our data suggest that AS-IV protected pancreatic β-cells from UA-treated dysfunction by activating the PI3K/AKT pathway. Such findings suggest that AS-IV may be an efficient natural agent against T2D.
Collapse
|
4
|
Katsiki N, Dimitriadis GD, Mikhailidis DP. Serum Uric Acid and Diabetes: From Pathophysiology to Cardiovascular Disease. Curr Pharm Des 2021; 27:1941-1951. [PMID: 33397230 DOI: 10.2174/1381612827666210104124320] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 11/13/2020] [Indexed: 11/22/2022]
Abstract
Hyperuricemia, has been traditionally related to nephrolithiasis and gout. However, it has also been associated with the development of type 2 diabetes mellitus (T2DM) and cardiometabolic and cardiovascular diseases. Pathophysiologically, elevated serum uric acid (SUA) levels may be associated with abnormal lipid and glucose metabolism. In this narrative review, we consider the associations between hyperuricemia, hyperglycemia, atherosclerosis and thrombosis. Furthermore, we comment on the available evidence linking elevated SUA levels with the incidence and outcomes of coronary heart disease, stroke, peripheral artery disease and non-alcoholic fatty liver in subjects with T2DM. The effects of antidiabetic drugs (e.g. metformin, pioglitazone, sulfonylureas, dipeptidyl peptidase 4 inhibitors, glucagon-like peptide-1 receptor agonists, sodium-glucose cotransporter 2 inhibitors and insulin) on SUA concentrations are also reviewed.
Collapse
Affiliation(s)
- Niki Katsiki
- First Department of Internal Medicine, Diabetes Center, Division of Endocrinology and Metabolism, AHEPA University Hospital, Thessaloniki, Greece
| | | | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital campus, University College London Medical School, University College London (UCL), London NW3 2QG, United Kingdom
| |
Collapse
|
5
|
Chen S, Yang F, Xu T, Wang Y, Zhang K, Fu G, Zhang W. Genetically predicted serum uric acid levels and the risk of coronary artery disease in patients with diabetes: A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2021; 31:1832-1839. [PMID: 33975736 DOI: 10.1016/j.numecd.2021.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Accepted: 03/07/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS Serum uric acid (SUA) levels have been reported to be associated with an increased risk of coronary artery disease (CAD) among patients with diabetes in observational study. Whether this relationship is causal remains unclear. The current study aimed to explore the causal association between SUA and the risk of CAD in patients with diabetes. METHODS AND RESULTS A two-sample Mendelian randomization (MR) approach was employed to evaluate the causal effect of SUA on the risk of CAD in patients with diabetes. A total of 28 single nucleotide polymorphisms (SNPs) related to SUA were identified as instruments. Genetic association with CAD were obtained from a recently published genome-wide association study (GWAS) of 15,666 patients with diabetes (3968 CAD cases and 11,696 controls). The fixed-effects inverse variance-weighted method was employed to estimate the causal effect for the primary analysis, and other robust methods were employed for sensitivity analyses. In addition, the whole analyses were repeated using 9 non-pleiotropic SNPs. Genetic determined SUA levels were not significantly associated with the risk of CAD in patients with diabetes in the primary analysis (odds ratio = 1.13, 95% confidence interval: 0.98-1.16, P = 0.09). Consistent results were observed in the sensitivity analyses using various robust methods. In addition, this finding was confirmed by the repeated analyses using 9 non-pleiotropic SNPs. CONCLUSIONS This two-sample MR study does not support a causal effect of genetically predicted SUA levels on the risk of CAD in patients with diabetes.
Collapse
Affiliation(s)
- Songzan Chen
- Department of Cardiology, Key laboratory of biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Fangkun Yang
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Tian Xu
- Department of Cardiology, Key laboratory of biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Yao Wang
- Department of Cardiology, Key laboratory of biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Kaijie Zhang
- Department of Cardiology, Key laboratory of biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Guosheng Fu
- Department of Cardiology, Key laboratory of biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.
| | - Wenbin Zhang
- Department of Cardiology, Key laboratory of biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
6
|
Jayachandran M, Qu S. Harnessing hyperuricemia to atherosclerosis and understanding its mechanistic dependence. Med Res Rev 2020; 41:616-629. [PMID: 33084092 DOI: 10.1002/med.21742] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/12/2020] [Accepted: 10/04/2020] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is regarded as the disease of the arterial vasculature. The main characteristics of atherosclerosis are the abnormal accumulation of lipids, increased inflammatory cells, matrix deposits, and proliferation of smooth muscle cells. Diabetes mellitus, obesity, and hyperlipidemia are the most studied risk factors of atherosclerosis. One least studied risk factor is the uric acid (UA), a high UA in circulation is interlinked with many pathological processes. Several epidemiological studies suggest elevated UA levels as an essential biomarker in the forecast of several cardiovascular diseases. Available evidence claims that UA upholds the atherosclerosis process via disturbing lipid metabolism, reducing the nitric oxide synthesis in endothelial cells, promoting the proliferation of vascular smooth muscle cells, and overwhelms inflammation. In endothelial dysfunction and coronary artery lesions, UA is considered as an independent predictor. The updated studies on the involvement of hyperuricemia in atherosclerosis prove that treatment with xanthine oxidase (XO) inhibitors not just benefits the treatment of hyperuricemia but also reduces the burden of atherosclerosis to a greater extent. In this review, we highlight how the hyperuricemia affects vascular integrity, causes atherosclerosis, and the mechanism of action of XO inhibitors on atherosclerosis.
Collapse
Affiliation(s)
- Muthukumaran Jayachandran
- Department of Endocrinology and Metabolism, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
7
|
Liu H, Ma X, Wang X, Ma X, Mao Z, Zhu J, Chen F. Hsa_circ_0000345 regulates the cellular development of ASMCs in response to oxygenized low-density lipoprotein. J Cell Mol Med 2020; 24:11849-11857. [PMID: 32865338 PMCID: PMC7578870 DOI: 10.1111/jcmm.15801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/19/2020] [Accepted: 08/08/2020] [Indexed: 12/31/2022] Open
Abstract
The interaction between circRNAs and atherosclerosis has been extensively studied. However, more novel circRNAs need to be explored to help establish a perfect regulatory network. In the present research, hsa_circ_0000345 was demonstrated to regulate cellular development of oxygenized low‐density lipoprotein (ox‐LDL)‐treated aortic smooth muscle cells (ASMCs), which was closely related to the occurrence and progress of atherosclerosis. Ox‐LDL exposure remarkably decreased hsa_circ_0000345 expression in ASMCs. Transfection‐induced hsa_circ_0000345 overexpression activated cell viability (detected by an MTT assay) and restrained cellular apoptosis (analysed by flow cytometry) in the atherosclerosis cellular model. While down‐regulation of hsa_circ_0000345 reduced cell viability and promoted cell apoptosis. In addition, the data of the cell cycle distribution analysis and trans‐well assay indicated that cell cycle progression was arrested at the G1 phase while cell invasion was enhanced in ASMCs following treatment of ox‐LDL in the context of hsa_circ_0000345 OE plasmids. In addition, up‐regulation of hsa_circ_0000345 supported HIF‐1α at both the mRNA and protein level, and down‐regulation of hsa_circ_0000345 reduced HIF‐1α expression. Overall, the above findings revealed that hsa_circ_0000345 was a dramatic regulator of ASMCs proliferation, apoptosis and invasion in response to ox‐LDL treatment. Hsa_circ_0000345 was identified as a protector of cell viability during ox‐LDL induced cell development.
Collapse
Affiliation(s)
- Huifang Liu
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowen Ma
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Wang
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xirui Ma
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziming Mao
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhu
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengling Chen
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|