1
|
Dempsey B, Pereira da Silva B, Cruz LC, Vileigas D, Silva ARM, Pereira da Silva R, Meotti FC. Unraveling the effects of uric acid on endothelial cells: A global proteomic study. Redox Biol 2025; 82:103625. [PMID: 40203480 PMCID: PMC12005352 DOI: 10.1016/j.redox.2025.103625] [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: 01/28/2025] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
This work aims to understand how normouricemic levels of uric acid can induce endothelial dysfunction seeking global proteomic alterations in Human Umbilical Vein cells (HUVEC). It reveals significant alterations in redox-sensitive and antioxidant proteins, chaperones, and proteins associated with cell migration and adhesion in response to uric acid exposure. Monitoring cellular oxidation with the roGFP2-Grx1 probe proved increased oxidation levels induced by uric acid, which can be attenuated by peroxidasin (PXDN) inhibition, suggesting a regulatory role for PXDN in mitigating oxidative stress induced by uric acid. As a consequence of uric acid oxidation and the formation of reactive intermediate, we identified adducts in proteins (+140 kDa) in a novel post-translation modification named uratylation. Increased misfolded protein levels and p62 aggregation were also found, indicating disturbances in cellular proteostasis. Furthermore, uric acid promoted monocyte adhesion and upregulated ICAM and VCAM protein levels, implicating a pro-inflammatory response in endothelial cells. These findings provide critical insights into the molecular mechanisms underlying vascular damage associated with uric acid.
Collapse
Affiliation(s)
- Bianca Dempsey
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Litiele Cezar Cruz
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Danielle Vileigas
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Amanda R M Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Flavia Carla Meotti
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
2
|
Waheed YA, Liu J, Almayahe S, Sun D. The role of hyperuricemia in the progression of end-stage kidney disease and its molecular prospective in inflammation and cardiovascular diseases: A general review. Ther Apher Dial 2025. [PMID: 39966090 DOI: 10.1111/1744-9987.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/15/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
With the ongoing development of the Chinese economy, the occurrence of chronic kidney disease (CKD) has experienced a remarkable upsurge recently, and due to uremia caused by CKD, the number of patients undergoing dialysis has shown a dramatic increase. China has been ranked first in the world for patients undergoing hemodialysis (HD) and peritoneal dialysis (PD) with approximately one million patients across the country. Due to the loss of kidney function caused by CKD, the kidneys tend to lose their ability to excrete uric acid (UA) out of the body; therefore, most patients undergoing dialysis are complicated with hyperuricemia (HUA). HUA is an abnormal disease of purine metabolism, and it's considered a chronic disease. More than 90% of patients suffering from HUA will not show any symptoms on physical examination. According to statistics, if high serum UA is left untreated, 55% of patients will develop severe problems due to the purine crystallization in the body, and the kidneys are the most affected organs by HUA causing renal insufficiency that can promote end-stage kidney disease (ESKD) by activating the renin-angiotensin system (RAS), which will lead to inflammation, arteriosclerosis, cardiovascular diseases (CVD), and other diseases. Lifestyle modifications and pharmacological interventions are the first primary choice for lowering UA, although dialysis will tend to reduce the high UA levels in the blood, drugs are also necessary. This review will summarize the mechanisms and metabolism of UA, the relationship between HUA and ESKD progression, HUA and inflammation, HUA and CVD, and pharmacological treatment of HUA.
Collapse
Affiliation(s)
- Yousuf Abdulkarim Waheed
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Clinical Research Center for Kidney Disease Xuzhou Medical University, Xuzhou, China
| | - Jie Liu
- Department of Nephrology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | | | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Clinical Research Center for Kidney Disease Xuzhou Medical University, Xuzhou, China
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Liu Y, Li Z, Xu Y, Mao H, Huang N. Uric Acid and Atherosclerosis in Patients with Chronic Kidney Disease: Recent Progress, Mechanisms, and Prospect. KIDNEY DISEASES (BASEL, SWITZERLAND) 2025; 11:112-127. [PMID: 40124130 PMCID: PMC11928073 DOI: 10.1159/000543781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/16/2025] [Indexed: 03/25/2025]
Abstract
Background Chronic kidney disease (CKD) is a prevalent global health concern, significantly linked to increased cardiovascular morbidity and mortality. Among various risk factors, uric acid (UA) has emerged as a potentially modifiable contributor to cardiovascular complications in CKD patients. Summary Elevated serum uric acid levels frequently occur in individuals with CKD and are associated with the development of atherosclerosis (AS). Uric acid has been demonstrated to exacerbate inflammatory processes, promote oxidative stress, and cause endothelial dysfunction, which are critical factors that drive the formation of atherosclerotic plaques. Furthermore, high uric acid levels can worsen renal function, establishing a detrimental cycle that amplifies cardiovascular risk. Key Messages This review investigates the complex interconnection between UA and AS in patients with CKD, highlighting the underlying mechanisms and therapeutic considerations. A more profound comprehension of this relationship is essential for enhancing cardiovascular health and outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Yuchu Liu
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Zeyu Li
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yuanwen Xu
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Naya Huang
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| |
Collapse
|
4
|
Xu J, Du X, Zhang S, Zang X, Xiao Z, Su R, Huang X, Liu L. Diagnostic value of uric acid to high-density lipoprotein cholesterol ratio in abdominal aortic aneurysms. Ann Med 2024; 56:2357224. [PMID: 38779715 PMCID: PMC11123539 DOI: 10.1080/07853890.2024.2357224] [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] [Received: 02/22/2024] [Accepted: 04/05/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is highly lethal upon onset of acute aortic diseases (AAD) or rupture. Dyslipidaemia and hyperuricaemia are important risk factors for the development of AAA and AAD as well as aortic disease-related death. The aim of this study was to explore whether uric acid (UA) to high-density lipoprotein cholesterol (HDL-C) ratio (UHR) can be used as an independent predictor of the presence of AAA or AAD. METHODS Three hundred subjects, including 100 AAA patients (AAA group), 100 AAD patients (AAD group) and 100 controls (CON group), were recruited in this study. UHR and other serum samples were obtained upon the patients' admission before any medical treatment. The optimal cut-off points of UHR were determined using receiver operating characteristic (ROC) curve analysis. RESULTS The UHR in AAA group was significantly higher than that in CON group, but there was no significant difference between AAD group and CON group. The optimal cut-off point of UHR for AAA was 7.78 (sensitivity 84.7%, specificity 62.4%, and AUC 0.811; p < 0.001), and UHR (OR: 1.122, 95%CI: 1.064-1.184; p < 0.001) was found to be an independent factor for predicting AAA after adjusting for traditional AAA risk factor. CONCLUSION UHR can be widely used in clinical practice as an auxiliary tool for screening AAA. The optimal cut-off point for UHR to AAA was determined for the first time in Chinese subjects.
Collapse
Affiliation(s)
- Jin Xu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Hunan, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Hunan, China
- Cardiovascular Disease Research Center of Hunan Province, Hunan, China
| | - Xiao Du
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shilan Zhang
- Department of Cardiovascular Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine
| | - Xueyan Zang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Hunan, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Hunan, China
- Cardiovascular Disease Research Center of Hunan Province, Hunan, China
| | - Zixi Xiao
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Hunan, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Hunan, China
- Cardiovascular Disease Research Center of Hunan Province, Hunan, China
| | - Rongkai Su
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Hunan, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Hunan, China
- Cardiovascular Disease Research Center of Hunan Province, Hunan, China
| | - Xiadie Huang
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Hunan, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Hunan, China
- Cardiovascular Disease Research Center of Hunan Province, Hunan, China
| | - Ling Liu
- Department of Cardiovascular Medicine, the Second Xiangya Hospital, Central South University, Hunan, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Hunan, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Hunan, China
- Cardiovascular Disease Research Center of Hunan Province, Hunan, China
| |
Collapse
|
5
|
Wei H, Zhan L, Lv X, Lin Y, Zheng J, Yang W, Liu J, Sun J, Chen S. Gut commensal Parabacteroides distasonis exerts neuroprotective effects in acute ischemic stroke with hyperuricemia via regulating gut microbiota-gut-brain axis. J Transl Med 2024; 22:999. [PMID: 39501312 PMCID: PMC11539330 DOI: 10.1186/s12967-024-05800-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Hyperuricemia is considered as an independent risk factor for acute ischemic stroke (AIS), and some AIS patients are accompanied by an increase in serum uric acid. Recent studies have highlighted the important role of gut microbiota in both hyperuricemia and AIS, but there is little available data on the relationship between gut microbiota and the pathogenesis of AIS with hyperuricemia (HAS). METHODS Here we profiled the gut microbiota composition in 63 HAS patients and 269 non-HAS patients through 16s rRNA sequencing. Male rat with hyperuricemia were subjected to middle cerebral artery occlusion (MCAO) to establish HAS model and were then treated with Parabacteroides distasonis. Subsequently, the neurological deficit, pathological damages and blood-brain barrier disruption were evaluated. Moreover, the levels of ROS, inflammatory cytokines, NF-𝜿B pathway related protein, and vascular density markers were determined. RESULTS There were significant differences of gut microbiota composition between HAS patients and non-HAS patients, and a significant decrease in the abundance of Parabacteroides in HAS patients compared to non-HAS patients. Animal experiments showed that supplementation with P. distasonis increased beneficial commensal bacteria, significantly improved neurological deficits, pathological damages and BBB disruption, as well as reduced the level of serum uric acid in HAS rats. We further demonstrated that P. distasonis treatment decreased ROS level and increased SOD2 level, thereby reducing oxidative stress. Meanwhile, P. distasonis effectively inhibited NF-𝜿B signal pathway and reduced the production of inflammatory cytokines, including TNF-α and IL-1β, alleviating the inflammatory response. Notably, P. distasonis treatment increased the levels of vascular density markers including cluster of differentiation 31 (CD31) and alpha-smooth muscle actin (α-SMA), ameliorating vascular damage in HAS rats. CONCLUSIONS Together, these findings highlighted the important role of P. distasonis in the pathogenesis of HAS, and its mechanism was involved in the regulation of gut microbiota-gut-brain axis, which implied a novel strategy against HAS.
Collapse
Affiliation(s)
- Hongming Wei
- Department of Geriatrics, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Lu Zhan
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xinhuang Lv
- Department of Geriatrics, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yan Lin
- Department of Geriatrics, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jie Zheng
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenwen Yang
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jing Sun
- Department of Geriatrics, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Songfang Chen
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
6
|
Zhang Y, Lian Q, Nie Y, Zhao W. Causal relationship between serum uric acid and cardiovascular disease: A Mendelian randomization study. IJC HEART & VASCULATURE 2024; 54:101453. [PMID: 39411145 PMCID: PMC11473680 DOI: 10.1016/j.ijcha.2024.101453] [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: 05/22/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 10/19/2024]
Abstract
Background Observational studies have established an association between serum uric acid and cardiovascular disease (CVD). However, these studies are susceptible to uncontrolled confounders and reverse causality bias. To overcome these challenges, we employed a two-sample Mendelian randomization (MR) approach to investigate the causal link between serum uric acid and CVD. Methods We utilized Genome-wide association study (GWAS) data for serum uric acid and six CVD: coronary artery disease (CAD), hypertension, myocardial infarction (MI), heart failure (HF), angina, and coronary heart disease (CHD). MR analyses employed inverse variance weighting (IVW), MR-Egger, weighted median, and weighted model. Sensitivity analyses were conducted to assess result reliability, including Cochrane's Q test, MR-Egger intercept, MR-PRESSO, and the leave-one-out approach. Results IVW analysis revealed that a genetic predisposition to elevated serum uric acid levels significantly increases the risk of CVD, with higher odds ratios (ORs) observed for CAD (OR: 1.227; 95 % CI: 1.107-1.360, P = 0.0002), hypertension (OR: 1.318, 95 %CI: 1.184-1.466, P = 2.13E-06), MI (OR: 1.184, 95 %CI: 1.108-1.266, P = 2.13E-06), HF (OR: 1.158, 95 %CI: 1.066-1.258, P = 2.13E-06), angina (OR: 1.150, 95 %CI: 1.074-1.231, P = 0.0002) and CHD (OR: 1.170, 95 %CI: 1.072-1.276, P = 0.0005). Sensitivity analysis research results have robustness. Conclusion This MR study robustly demonstrates a significant causal relationship between genetically elevated serum uric acid and various cardiovascular diseases, suggesting that higher levels may enhance the risk of cardiovascular events. Consequently, patients with elevated uric acid levels warrant early and aggressive interventions to mitigate cardiovascular risks.
Collapse
Affiliation(s)
- Yujun Zhang
- Yan’an University Xianyang Hospital, Data Management Center, Xianyang, China
| | - Qiufang Lian
- Department of Cardiology, Xianyang Hospital, Yan’an University, Xianyang, China
| | - Yanwu Nie
- School of Public Health, Nanchang University, Nanchang, China
| | - Wei Zhao
- Department of Cardiology, Xianyang Hospital, Yan’an University, Xianyang, China
| |
Collapse
|
7
|
van der Pol KH, Koenderink J, van den Heuvel JJMW, van den Broek P, Peters J, van Bunningen IDW, Pertijs J, Russel FGM, Koldenhof J, Morshuis WJ, van Drongelen J, Schirris TJJ, van der Meer A, Rongen GA. Effects of allopurinol and febuxostat on uric acid transport and transporter expression in human umbilical vein endothelial cells. PLoS One 2024; 19:e0305906. [PMID: 38905201 PMCID: PMC11192402 DOI: 10.1371/journal.pone.0305906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024] Open
Abstract
Uric acid induces radical oxygen species formation, endothelial inflammation, and endothelial dysfunction which contributes to the progression of atherosclerosis. Febuxostat inhibits BCRP- and allopurinol stimulates MRP4-mediated uric acid efflux in human embryonic kidney cells. We hypothesized that endothelial cells express uric acid transporters that regulate intracellular uric acid concentration and that modulation of these transporters by febuxostat and allopurinol contributes to their different impact on cardiovascular mortality. The aim of this study was to explore a potential difference between the effect of febuxostat and allopurinol on uric acid uptake by human umbilical vein endothelial cells. Febuxostat increased intracellular uric acid concentrations compared with control. In contrast, allopurinol did not affect intracellular uric acid concentration. In line with this observation, febuxostat increased mRNA expression of GLUT9 and reduced MRP4 expression, while allopurinol did not affect mRNA expression of these uric acid transporters. These findings provide a possible pathophysiological pathway which could explain the higher cardiovascular mortality for febuxostat compared to allopurinol but should be explored further.
Collapse
Affiliation(s)
- Karel H. van der Pol
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan Koenderink
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Petra van den Broek
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janny Peters
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Imke D. W. van Bunningen
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeanne Pertijs
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans G. M. Russel
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jim Koldenhof
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
| | - Wim J. Morshuis
- Department of Cardio-thoracic Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joris van Drongelen
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom J. J. Schirris
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Gerard A. Rongen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
8
|
Lee SA, Riella LV. Narrative Review of Immunomodulatory and Anti-inflammatory Effects of Sodium-Glucose Cotransporter 2 Inhibitors: Unveiling Novel Therapeutic Frontiers. Kidney Int Rep 2024; 9:1601-1613. [PMID: 38899203 PMCID: PMC11184259 DOI: 10.1016/j.ekir.2024.02.1435] [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: 12/19/2023] [Revised: 02/08/2024] [Accepted: 02/25/2024] [Indexed: 06/21/2024] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2 inhibitors) have evolved from their initial role as antidiabetic drugs to garner recognition for their remarkable cardio-protective and reno-protective attributes. They have become a crucial component of therapeutic guidelines for congestive heart failure and proteinuric chronic kidney disease (CKD). These benefits extend beyond glycemic control, because improvements in cardiovascular and renal outcomes occur swiftly. Recent studies have unveiled the immunomodulatory properties of SGLT2 inhibitors; thus, shedding light on their potential to influence the immune system and inflammation. This comprehensive review explores the current state of knowledge regarding the impact of SGLT2 inhibitors on the immune system and inflammation, focusing on preclinical and clinical evidence. The review delves into their antiinflammatory and immunomodulating effects, offering insights into clinical implications, and exploring emerging research areas related to their prospective immunomodulatory impact.
Collapse
Affiliation(s)
- Sul A. Lee
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Leonardo V. Riella
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Medicine and Surgery, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Wen S, Arakawa H, Tamai I. Uric acid in health and disease: From physiological functions to pathogenic mechanisms. Pharmacol Ther 2024; 256:108615. [PMID: 38382882 DOI: 10.1016/j.pharmthera.2024.108615] [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: 12/28/2023] [Revised: 02/02/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024]
Abstract
Owing to renal reabsorption and the loss of uricase activity, uric acid (UA) is strictly maintained at a higher physiological level in humans than in other mammals, which provides a survival advantage during evolution but increases susceptibility to certain diseases such as gout. Although monosodium urate (MSU) crystal precipitation has been detected in different tissues of patients as a trigger for disease, the pathological role of soluble UA remains controversial due to the lack of causality in the clinical setting. Abnormal elevation or reduction of UA levels has been linked to some of pathological status, also known as U-shaped association, implying that the physiological levels of UA regulated by multiple enzymes and transporters are crucial for the maintenance of health. In addition, the protective potential of UA has also been proposed in aging and some diseases. Therefore, the role of UA as a double-edged sword in humans is determined by its physiological or non-physiological levels. In this review, we summarize biosynthesis, membrane transport, and physiological functions of UA. Then, we discuss the pathological involvement of hyperuricemia and hypouricemia as well as the underlying mechanisms by which UA at abnormal levels regulates the onset and progression of diseases. Finally, pharmacological strategies for urate-lowering therapy (ULT) are introduced, and current challenges in UA study and future perspectives are also described.
Collapse
Affiliation(s)
- Shijie Wen
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
10
|
Hu Q, Lan H, Tian Y, Li X, Wang M, Zhang J, Yu Y, Chen W, Kong L, Guo Y, Zhang Z. Biofunctional coacervate-based artificial protocells with membrane-like and cytoplasm-like structures for the treatment of persistent hyperuricemia. J Control Release 2024; 365:176-192. [PMID: 37992873 DOI: 10.1016/j.jconrel.2023.11.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023]
Abstract
Coacervate droplets formed by liquid-liquid phase separation have attracted considerable attention due to their ability to enrich biomacromolecules while preserving their bioactivities. However, there are challenges to develop coacervate droplets as delivery vesicles for therapeutics resulting from the lack of physiological stability and inherent lack of membranes in coacervate droplets. Herein, polylysine-polynucleotide complex coacervate droplets with favorable physiological stability are formulated to efficiently and facilely concentrate small molecules, biomacromolecules and nanoparticles without organic solvents. To improve the biocompatibility, the PEGylated phospholipid membrane is further coated on the surface of the coacervate droplets to prepare coacervate-based artificial protocells (ArtPC) with membrane-like and cytoplasm-like structures. The ArtPC can confine the cyclic catalytic system of uricase and catalase inside to degrade uric acid and deplete the toxicity of H2O2. This biofunctional ArtPC effectively reduces blood uric acid levels and prevents renal injuries in mice with persistent hyperuricemia. The ArtPC-based therapy can bridge the disciplines of synthetic biology, pharmaceutics and therapeutics.
Collapse
Affiliation(s)
- Qian Hu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongbing Lan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinmei Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaonan Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengmeng Wang
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiao Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yulin Yu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Chen
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuanyuan Guo
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Engineering Research Centre for Novel Drug Delivery System, Wuhan 430030, China.
| |
Collapse
|
11
|
Zhu Y, Di S, Li Y, Liang W, Liu J, Nuermaimaiti R, Fei W, Wang C, Wang L, Zhang J. Integrative metabolomic and network pharmacological analysis reveals potential mechanisms of Cardamine circaeoides Hook.f. & Thomson in alleviating potassium oxonate-induced asymptomatic hyperuricemia in rats. Front Pharmacol 2023; 14:1281411. [PMID: 38026974 PMCID: PMC10652788 DOI: 10.3389/fphar.2023.1281411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Cardamine circaeoides Hook.f. & Thomson (CC), a herb of the genus Cardamine (family Brassicaceae), has a rich historical usage in China for both culinary and medicinal purposes. It is distinguished by its remarkable ability to hyperaccumulate selenium (Se). CC has demonstrated efficacy in the prevention of metabolic disorders. However, investigations into the effects of CC on asymptomatic hyperuricemia remain scarce. The objective of this study is to elucidate the mechanism by which CC aqueous extract (CCE) exerts its anti-hyperuricemic effects on asymptomatic hyperuricemic rats induced by potassium oxonate (PO) by integrating metabolomics and network pharmacological analysis. Asymptomatic hyperuricemia was induced by feeding rats with PO (1000 mg/kg) and CCE (0.75, 1.5, or 3 g/kg) once daily for 30 days. Various parameters, including body weight, uric acid (UA) levels, histopathology of renal tissue, and inflammatory factors (IL-1β, IL-6, IL-8, and TNF-α) were assessed. Subsequently, metabolomic analysis of kidney tissues was conducted to explore the effects of CCE on renal metabolites and the related pathways. Furthermore, network pharmacology was employed to explicate the mechanism of action of CCE components identified through UPLC-Q-TOF-MS analysis. Finally, metabolomic and network-pharmacology analyses were performed to predict crucial genes dysregulated in the disease model and rescued by CCE, which were then subjected to verification by RT-qPCR. The findings revealed that CCE significantly inhibited the UA levels from the 21st day to the 30th day. Moreover, CCE exhibited significant inhibition of IL-1β, IL-6, IL-8, and TNF-α levels in renal tissues. The dysregulation of 18 metabolites and the tyrosine, pyrimidine, cysteine, methionine, sphingolipid, and histidine metabolism pathways was prevented by CCE treatment. A joint analysis of targets predicted using the network pharmacology approach and the differential metabolites found in metabolics predicted 8 genes as potential targets of CCE, and 3 of them (PNP gene, JUN gene, and ADA gene) were verified at the mRNA level by RT-qPCR. We conclude that CCE has anti-hyperuricemia effects and alleviates renal inflammation in a rat model of hyperuricemia, and these efficacies are associated with the reversal of increased ADA, PNP, and JUN mRNA expression in renal tissues.
Collapse
Affiliation(s)
- Yingli Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Songrui Di
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yipeng Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weican Liang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jinlian Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Reyisai Nuermaimaiti
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenting Fei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chun Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Linyuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
12
|
Tanaka Y, Nagoshi T, Takahashi H, Oi Y, Yasutake R, Yoshii A, Kimura H, Kashiwagi Y, Tanaka TD, Shimoda M, Yoshimura M. URAT1 is expressed in cardiomyocytes and dotinurad attenuates the development of diet-induced metabolic heart disease. iScience 2023; 26:107730. [PMID: 37694143 PMCID: PMC10483053 DOI: 10.1016/j.isci.2023.107730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/17/2023] [Accepted: 08/23/2023] [Indexed: 09/12/2023] Open
Abstract
We recently reported that the selective inhibition of urate transporter-1 (URAT1), which is primarily expressed in the kidneys, ameliorates insulin resistance by attenuating hepatic steatosis and improving brown adipose tissue function in diet-induced obesity. In this study, we evaluated the effects of dotinurad, a URAT1-selective inhibitor, on the hearts of high-fat diet (HFD)-fed obese mice for 16-20 weeks and on neonatal rat cardiomyocytes (NRCMs) exposed to palmitic acid. Outside the kidneys, URAT1 was also expressed in cardiomyocytes and indeed worked as a uric acid transporter. Dotinurad substantially attenuated HFD-induced cardiac fibrosis, inflammatory responses, and cardiac dysfunction. Intriguingly, among various factors related to the pathophysiology of diet-induced obesity, palmitic acid significantly increased URAT1 expression in NRCMs and subsequently induced apoptosis, oxidative stress, and inflammatory responses via MAPK pathway, all of which were reduced by dotinurad. These results indicate that URAT1 is a potential therapeutic target for metabolic heart disease.
Collapse
Affiliation(s)
- Yoshiro Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hirotake Takahashi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yuhei Oi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Rei Yasutake
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Akira Yoshii
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yusuke Kashiwagi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Toshikazu D. Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
13
|
Zhang WZ. Uric acid en route to gout. Adv Clin Chem 2023; 116:209-275. [PMID: 37852720 DOI: 10.1016/bs.acc.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Gout and hyperuricemia (HU) have generated immense attention due to increased prevalence. Gout is a multifactorial metabolic and inflammatory disease that occurs when increased uric acid (UA) induce HU resulting in monosodium urate (MSU) crystal deposition in joints. However, gout pathogenesis does not always involve these events and HU does not always cause a gout flare. Treatment with UA-lowering therapeutics may not prevent or reduce the incidence of gout flare or gout-associated comorbidities. UA exhibits both pro- and anti-inflammation functions in gout pathogenesis. HU and gout share mechanistic and metabolic connections at a systematic level, as shown by studies on associated comorbidities. Recent studies on the interplay between UA, HU, MSU and gout as well as the development of HU and gout in association with metabolic syndromes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular, renal and cerebrovascular diseases are discussed. This review examines current and potential therapeutic regimens and illuminates the journey from disrupted UA to gout.
Collapse
Affiliation(s)
- Wei-Zheng Zhang
- VIDRL, The Peter Doherty Institute, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Liu P, Ma G, Wang Y, Wang L, Li P. Therapeutic effects of traditional Chinese medicine on gouty nephropathy: Based on NF-κB signalingpathways. Biomed Pharmacother 2023; 158:114199. [PMID: 36916428 DOI: 10.1016/j.biopha.2022.114199] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/20/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
As the final product of purine metabolism, excess serum uric acid (SUA) aggravates the process of some metabolic diseases. SUA causes renal tubule damage, interstitial fibrosis, and glomerular hardening, leading to gouty nephropathy (GN). A growing number of investigations have shown that NF-κB mediated inflammation and oxidative stress have been directly involved in the pathogenesis of GN. Traditional Chinese medicine's treatment methods of GN have amassed a wealth of treatment experience. In this review, we first describe the mechanism of NF-κB signaling pathways in GN. Subsequently, we highlight traditional Chinese medicine that can treat GN through NF-κB pathways. Finally, commenting on promising candidate targets of herbal medicine for GN treatment via suppressing NF-κB signaling pathways was summarized.
Collapse
Affiliation(s)
- Peng Liu
- Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Station East 5, Shunyi District, Beijing 101300, China
| | - Guijie Ma
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Yang Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Lifan Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China.
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
15
|
Cimmino G, Conte S, Marra L, Morello A, Morello M, De Rosa G, Pepe M, Sugraliyev A, Golino P, Cirillo P. Uric Acid Induces a Proatherothrombotic Phenotype in Human Endothelial Cells by Imbalancing the Tissue Factor/Tissue Factor Pathway Inhibitor Pathway. Thromb Haemost 2023; 123:64-75. [PMID: 36126947 DOI: 10.1055/a-1947-7716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Several evidence show that elevated plasma levels of uric acid (UA) are associated with the increased risk of developing atherothrombotic cardiovascular events. Hyperuricemia is a risk factor for endothelial dysfunction (ED). ED is involved in the pathophysiology of atherothrombosis since dysfunctional cells lose their physiological, antithrombotic properties. We have investigated whether UA might promote ED by modulating the tissue factor (TF)/TF pathway inhibitor (TFPI) balance by finally changing the antithrombotic characteristics of endothelial cells. METHODS Human umbilical vein endothelial cells were incubated with increasing doses of UA (up to 9 mg/dL). TF gene and protein expressions were evaluated by real-time polymerase chain reaction (PCR) and Western blot. Surface expression and procoagulant activity were assessed by FACS (fluorescence activated cell sorting) analysis and coagulation assay. The mRNA and protein levels of TFPI were measured by real-time PCR and Western blot. The roles of inflammasome and nuclear factor-κB (NF-κB) as possible mechanism(s) of action of the UA on TF/TFPI balance were also investigated. RESULTS UA significantly increased TF gene and protein levels, surface expression, and procoagulant activity. In parallel, TFPI levels were significantly reduced. The NF-κB pathways appeared to be involved in modulating these phenomena. Additionally, inflammasome might also play a role. CONCLUSION The present in vitro study shows that one of the mechanisms by which high levels of UA contribute to ED might be the imbalance between TF/TFPI levels in endothelial cells, shifting them to a nonphysiological, prothrombotic phenotype. These UA effects might hypothetically explain, at least in part, the relationship observed between elevated plasma levels of UA and cardiovascular events.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Section of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Stefano Conte
- Section of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Laura Marra
- SC Cell Biology and Biotherapy, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Andrea Morello
- Biochemical Unit, Azienda Sanitaria Regionale Molise, Antonio Cardarelli Hospital, Campobasso, Italy
| | - Mariarosaria Morello
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples "Federico II," Naples, Italy
| | - Gennaro De Rosa
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples "Federico II," Naples, Italy
| | - Martino Pepe
- Cardiovascular Diseases Section, Department of Emergency and Organ Transplantation (DETO), University of Bari, Bari, Italy
| | - Akhmetzhan Sugraliyev
- Department of Internal Disease, Kazakh National Medical University, Almaty, Kazakhstan
| | - Paolo Golino
- Section of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Plinio Cirillo
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples "Federico II," Naples, Italy
| |
Collapse
|
16
|
Vankova D, Kiselova-Kaneva Y, Ivanova D. Uric acid effects on glutathione metabolism estimated by induction of glutamate-cysteine ligase, glutathione reductase and glutathione synthetase in mouse J744A.1 macrophage cell line. Folia Med (Plovdiv) 2022; 64:762-769. [PMID: 36876528 DOI: 10.3897/folmed.64.e65507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/28/2021] [Indexed: 11/07/2022] Open
Abstract
INTRODUCTION Elevated plasma levels of uric acid (UA) are considered an independent risk factor for hypertension, diabetes, cardiovascular disease, endothelial and vascular damage, obesity, and metabolic syndrome. Even physiological concentrations of soluble UA have been proved to induce gene expression of macrophage-secreted inflammatory cytokines and stimulate production of reactive oxygen species in mature adipocytes. UA is also described as a powerful endogenous plasma antioxidant, which reveals a paradox of duality for this parameter.
Collapse
|
17
|
Nardi V, Franchi F, Prasad M, Fatica EM, Alexander MP, Bois MC, Lam J, Singh RJ, Meyer FB, Lanzino G, Xiong Y, Lutgens E, Lerman LO, Lerman A. Uric Acid Expression in Carotid Atherosclerotic Plaque and Serum Uric Acid Are Associated With Cerebrovascular Events. Hypertension 2022; 79:1814-1823. [PMID: 35656807 DOI: 10.1161/hypertensionaha.122.19247] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Uric acid (UA) concentration within carotid plaque and its association with cerebrovascular events have not been detected or quantified. Systemically, serum UA is a marker of inflammation and risk factor for atherosclerosis. However, its association with carotid plaque instability and stroke pathogenesis remains unclear. In patients undergoing carotid endarterectomy, we aimed to determine whether UA is present differentially in symptomatic versus asymptomatic carotid plaques and whether serum UA is associated with cerebrovascular symptoms (stroke, transient ischemic attack, or amaurosis fugax). METHODS Carotid atherosclerotic plaques were collected during carotid endarterectomy. The presence of UA was assessed using Gomori methenamine silver staining as well as anti-UA immunohistochemical staining and its quantity measured using an enzymatic colorimetric assay. Clinical information was obtained through a retrospective review of data. RESULTS UA was more commonly detected in symptomatic (n=23) compared with asymptomatic (n=9) carotid plaques by Gomori methenamine silver (20 [86.9%] versus 2 [22.2%]; P=0.001) and anti-UA immunohistochemistry (16 [69.5%] versus 1 [11.1%]; P=0.004). UA concentration was higher in symptomatic rather than asymptomatic plaques (25.1 [9.5] versus 17.9 [3.8] µg/g; P=0.021). Before carotid endarterectomy, serum UA levels were higher in symptomatic (n=341) compared with asymptomatic (n=146) patients (5.9 [interquartile range, 4.6-6.9] mg/dL versus 5.2 [interquartile range, 4.6-6.2] mg/dL; P=0.009). CONCLUSIONS The current study supports a potential role of UA as a potential tissue participant and a systemic biomarker in the pathogenesis of carotid atherosclerosis. UA may provide a mechanistic explanation for plaque instability and subsequent ischemic cerebrovascular events.
Collapse
Affiliation(s)
- Valentina Nardi
- Department of Cardiovascular Medicine (V.N., F.F., M.P., J.L., Y.X., E.L., A.L.), Mayo Clinic, Rochester, MN
| | - Federico Franchi
- Department of Cardiovascular Medicine (V.N., F.F., M.P., J.L., Y.X., E.L., A.L.), Mayo Clinic, Rochester, MN
| | - Megha Prasad
- Department of Cardiovascular Medicine (V.N., F.F., M.P., J.L., Y.X., E.L., A.L.), Mayo Clinic, Rochester, MN
| | - Erica M Fatica
- Department of Laboratory of Medicine and Pathology (E.M.F., M.P.A., M.C.B., R.J.S.), Mayo Clinic, Rochester, MN
| | - Mariam P Alexander
- Department of Laboratory of Medicine and Pathology (E.M.F., M.P.A., M.C.B., R.J.S.), Mayo Clinic, Rochester, MN
| | - Melanie C Bois
- Department of Laboratory of Medicine and Pathology (E.M.F., M.P.A., M.C.B., R.J.S.), Mayo Clinic, Rochester, MN
| | - Josephine Lam
- Department of Cardiovascular Medicine (V.N., F.F., M.P., J.L., Y.X., E.L., A.L.), Mayo Clinic, Rochester, MN
| | - Ravinder J Singh
- Department of Laboratory of Medicine and Pathology (E.M.F., M.P.A., M.C.B., R.J.S.), Mayo Clinic, Rochester, MN
| | - Fredric B Meyer
- Department of Neurosurgery (F.B.M., G.L.), Mayo Clinic, Rochester, MN
| | - Giuseppe Lanzino
- Department of Neurosurgery (F.B.M., G.L.), Mayo Clinic, Rochester, MN
| | - Yuning Xiong
- Department of Cardiovascular Medicine (V.N., F.F., M.P., J.L., Y.X., E.L., A.L.), Mayo Clinic, Rochester, MN
| | - Esther Lutgens
- Department of Cardiovascular Medicine (V.N., F.F., M.P., J.L., Y.X., E.L., A.L.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- Department of Nephrology and Hypertension (L.O.L.), Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Department of Cardiovascular Medicine (V.N., F.F., M.P., J.L., Y.X., E.L., A.L.), Mayo Clinic, Rochester, MN
| |
Collapse
|
18
|
Dempsey B, Cruz LC, Mineiro MF, da Silva RP, Meotti FC. Uric Acid Reacts with Peroxidasin, Decreases Collagen IV Crosslink, Impairs Human Endothelial Cell Migration and Adhesion. Antioxidants (Basel) 2022; 11:antiox11061117. [PMID: 35740014 PMCID: PMC9220231 DOI: 10.3390/antiox11061117] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Uric acid is considered the main substrate for peroxidases in plasma. The oxidation of uric acid by human peroxidases generates urate free radical and urate hydroperoxide, which might affect endothelial function and explain, at least in part, the harmful effects of uric acid on the vascular system. Peroxidasin (PXDN), the most recent heme-peroxidase described in humans, catalyzes the formation of hypobromous acid, which mediates collagen IV crosslinks in the extracellular matrix. This enzyme has gained increasing scientific interest since it is associated with cardiovascular disease, cancer, and renal fibrosis. The main objective here was to investigate whether uric acid would react with PXDN and compromise the function of the enzyme in human endothelial cells. Urate decreased Amplex Red oxidation and brominating activity in the extracellular matrix (ECM) from HEK293/PXDN overexpressing cells and in the secretome of HUVECs. Parallelly, urate was oxidized to 5-hydroxyisourate. It also decreased collagen IV crosslink in isolated ECM from PFHR9 cells. Urate, the PXDN inhibitor phloroglucinol, and the PXDN knockdown impaired migration and adhesion of HUVECs. These results demonstrated that uric acid can affect extracellular matrix formation by competing for PXDN. The oxidation of uric acid by PXDN is likely a relevant mechanism in the endothelial dysfunction related to this metabolite.
Collapse
|
19
|
Wei C, Yu X, Wang L, Jiang J, Dai Q, Kang Y, Li J, Chen X. Can hyperuricemia predict the progression risk of cerebral small vessel disease? Neurol Res 2022; 44:910-917. [PMID: 35475780 DOI: 10.1080/01616412.2022.2067707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIMS Uric acid (UA) may play a crucial role in the process of cerebral small vessel disease (SVD), but few follow-up studies have focused on the effect of UA in the progression of SVD. The present study aimed to ascertain whether serum UA levels are associated with the risk of SVD progression. METHODS We performed an observational clinical study in adults older than 45 years with cranial magnetic resonance imaging (MRI) from 30 October 2015, to 28 January 2021. The patients were divided into two groups according to whether their total burden of SVD scores increased or not during the follow-up: SVD progression (increased by at least one point) and without SVD progression (increased 0 points). Cox regression and Kaplan-Meier survival analyses were used for univariate analysis between groups to identify the risk factors for SVD progression. RESULTS Ultimately, 261 eligible patients were included in the final analysis. Of the 261 eligible patients, 73 were included in the SVD progression group, and 188 were included in the group without SVD progression. Correlation analysis found that the levels of UA and the ratio of hyperuricemia (HUA) showed statistically significant correlations with SVD progression risk (r = 0.197 and Crammer's V = 0.213, respectively, P < 0.01). Cox regression and Kaplan-Meier survival analyses showed that after adjustment for covariates, HUA was an independent risk factor for the incidence of SVD progression. The risk of SVD progression in patients with HUA was higher than that in those without HUA (HR (95% CI), 1.77 (1.03-3.05), P < 0.05). CONCLUSIONS High serum UA levels are independently related to the risk of SVD progression, thus highlighting not only the influence of traditional risk factors such as hypertension and age on SVD but also the UA levels of patients for individualized treatment.
Collapse
Affiliation(s)
- Cunsheng Wei
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaorong Yu
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lin Wang
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junying Jiang
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qi Dai
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Kang
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junrong Li
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuemei Chen
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
20
|
Liang X, Liu X, Li D, Qin W, Liu Y. Effectiveness of Urate-Lowering Therapy for Renal Function in Patients With Chronic Kidney Disease: A Meta-Analysis of Randomized Clinical Trials. Front Pharmacol 2022; 13:798150. [PMID: 35370725 PMCID: PMC8968869 DOI: 10.3389/fphar.2022.798150] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/24/2022] [Indexed: 02/05/2023] Open
Abstract
Background and Objective: Hyperuricemia is closely related to chronic kidney disease (CKD). The effects of urate-lowering therapy (ULT) on renal outcomes are uncertain, and whether it is warranted in CKD patients is currently unclear. The aim of our meta-analysis of randomized clinical trials (RCTs) was to assess the effectiveness and safety of ULT for improving kidney function in patients with CKD. Methods: RCTs were retrieved from the PubMed, Embase, MEDLINE and Cochrane Central Register of Controlled Trials databases. The meta-analysis was performed using Review Manager and Stata/SE software. The outcomes were changes in renal function and serum uric acid (SUA), serum creatinine, and adverse events. Results: Twelve RCTs with 1,469 participants were included in the meta-analysis. ULT was found to effectively lower SUA (standard mean difference (SMD): -2.70; 95% confidence interval (CI): -3.71, -1.69) but the renoprotective effects were not superior to those of control therapy (placebo or usual therapy), which were stable in the subgroup analyses and sensitivity analyses. Regarding adverse events, their risks did not increase in the ULT group compared with the control group and were stable in the sensitivity analyses. Conclusion: The findings of our meta-analysis suggested that ULT can effectively lower SUA, but there is insufficient evidence to support the renoprotective effects of ULT in CKD patients. In addition, ULT is safe for patients with CKD. Systematic Review Registration:https://clinicaltrials.gov/, identifier PROSPERO (CRD42020200550).
Collapse
Affiliation(s)
- Xiuping Liang
- Department of Rheumatology and Immunology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiang Liu
- Department of Nephrology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| | - Duohui Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Wei Qin
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Ng HY, Leung FF, Kuo WH, Lee WC, Lee CT. Dapagliflozin and xanthine oxidase inhibitors improve insulin resistance and modulate renal glucose and urate transport in metabolic syndrome. Clin Exp Pharmacol Physiol 2021; 48:1603-1612. [PMID: 34407232 DOI: 10.1111/1440-1681.13574] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 11/28/2022]
Abstract
Disturbance in glucose and uric acid metabolism is the major disorder of metabolic syndrome (MetS). The kidneys play an important role in the management of glucose and uric acid. The aim of our study was to investigate alterations in renal glucose and uric acid transporters in animals with MetS after treatment with dapagliflozin and xanthine oxidase inhibitors (allopurinol and febuxostat). Sprague-Dawley rats were fed normal chow or a high fructose diet for the first 3 months. The fructose-fed animals were then treated with dapagliflozin, allopurinol, febuxostat, or no treatment for the next 3 months. Fasting glucose, insulin resistance, and hyperuricaemia were improved in all treatment groups except that in the fructose group (all p < 0.05). Both allopurinol and febuxostat reversed the increase in levels of sodium glucose cotransporter (SGLT) 1, SGLT2, and glucose transporter (GLUT) 2 (all p < 0.05). Dapagliflozin alleviated hyperuricaemia and induced uricosuria without affecting serum xanthine oxidase activity. Dapagliflozin suppressed the expression of GLUT9, urate transporter, and urate anion exchanger 1 (all p < 0.05), which was similar to the effects of allopurinol and febuxostat. The results suggest that treatment with dapagliflozin and xanthine oxidase inhibitors improved insulin resistance and reversed the increased expression of glucose and urate transporters in the kidney.
Collapse
Affiliation(s)
- Hwee-Yeong Ng
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Foong-Fah Leung
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wei-Hung Kuo
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Chin Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
22
|
Kang EH, Park EH, Shin A, Song JS, Kim SC. Cardiovascular risk associated with allopurinol vs. benzbromarone in patients with gout. Eur Heart J 2021; 42:4578-4588. [PMID: 34508567 PMCID: PMC8633759 DOI: 10.1093/eurheartj/ehab619] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/06/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
Aims With the high prevalence of gout and associated cardiovascular (CV) diseases, information on the comparative CV safety of individual urate-lowering drugs becomes increasingly important. However, few studies examined the CV risk of uricosuric agents. We compared CV risk among patients with gout who initiated allopurinol vs. benzbromarone. Methods and results Using the Korean National Health Insurance claims data (2002–17), we conducted a cohort study of 124 434 gout patients who initiated either allopurinol (n = 103 695) or benzbromarone (n = 20 739), matched on propensity score at a 5:1 ratio. The primary outcome was a composite CV endpoint of myocardial infarction, stroke/transient ischaemic attack, or coronary revascularization. To account for competing risk of death, we used cause-specific hazard models to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for the outcomes comparing allopurinol initiators with benzbromarone. Over a mean follow-up of 1.16 years, 2258 patients developed a composite CV event. The incidence rate of the composite CV event was higher in allopurinol initiators (1.81 per 100 person-years) than benzbromarone (1.61 per 100 person-years) with a HR of 1.22 (95% CI 1.05–1.41). The HR for all-cause mortality was 1.66 (95% CI 1.43–1.93) among allopurinol initiators compared with benzbromarone. Conclusion In this large population-based cohort of gout patients, allopurinol was associated with an increased risk of composite CV events and all-cause mortality compared to benzbromarone. Benzbromarone may reduce CV risk and mortality in patients with gout, although more studies are necessary to confirm our findings and to advance our understanding of the underlying mechanisms.
Collapse
Affiliation(s)
- Eun Ha Kang
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, 166 Gumiro Bundang-gu, Seongnam, South Korea
| | - Eun Hye Park
- Division of Rheumatology, Department of Internal Medicine, Chung-Ang University, School of Medicine, Seoul, South Korea
| | - Anna Shin
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, 166 Gumiro Bundang-gu, Seongnam, South Korea
| | - Jung Soo Song
- Division of Rheumatology, Department of Internal Medicine, Chung-Ang University, School of Medicine, Seoul, South Korea
| | - Seoyoung C Kim
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Nie Q, Liu M, Zhang Z, Zhang X, Wang C, Song G. The effects of hyperuricemia on endothelial cells are mediated via GLUT9 and the JAK2/STAT3 pathway. Mol Biol Rep 2021; 48:8023-8032. [PMID: 34716863 PMCID: PMC8604859 DOI: 10.1007/s11033-021-06840-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/12/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Uric acid (UA) transporters mediate the uptake and outflow of UA, and are greatly involved in the control of UA concentrations. Glucose transporter 9 (GLUT9), one of the UA transporters, has been confirmed to be expressed in human umbilical vein endothelial cells (HUVECs). This study aimed to characterize GLUT9's effect on intracellular UA accumulation in HUVECs in a high-UA environment and to explore the mechanism of cellular dysfunction. METHODS AND RESULTS HUVECs were treated with UA to establish a model of cellular dysfunction. Then, UA uptake, GLUT9 expression and endothelial nitric oxide synthase (eNOS) and reactive oxygen species (ROS) amounts were measured. UA uptake was concentration- and time-dependent, and UA treatment significantly reduced nitric oxide (NO) levels and eNOS activity. UA also upregulated pro-inflammatory molecules and GLUT9, and increased intracellular ROS amounts in HUVECs. GLUT9 knockdown reduced UA uptake and ROS content, but antioxidant treatment did not reduce GLUT9 expression. To assess the function of JAK2/STAT3 signaling, HUVECs were treated with UA, and the phosphorylation levels of JAK2, STAT3, IL-6 and SOCS3 were increased by a high concentration of UA. In addition, GLUT9 knockdown reduced the phosphorylation of JAK2/STAT3 intermediates and increased p-eNOS amounts. CONCLUSIONS GLUT9 mediated the effects of high UA levels on HUVECs by increasing the cellular uptake of UA, activating JAK2/STAT3 signaling, and reduced the production of active eNOS and NO in HUVECs.
Collapse
Affiliation(s)
- Qian Nie
- Department of Internal Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China.,Physical Examination Center of Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Miaomiao Liu
- Department of Oncology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Zhimei Zhang
- Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Xuemei Zhang
- Department of Rheumatism and Immunology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Chao Wang
- Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Guangyao Song
- Department of Internal Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China. .,Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China.
| |
Collapse
|
24
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Hyperuricemia-induced endothelial insulin resistance: the nitric oxide connection. Pflugers Arch 2021; 474:83-98. [PMID: 34313822 DOI: 10.1007/s00424-021-02606-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/12/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022]
Abstract
Hyperuricemia, defined as elevated serum concentrations of uric acid (UA) above 416 µmol L-1, is related to the development of cardiometabolic disorders, probably via induction of endothelial dysfunction. Hyperuricemia causes endothelial dysfunction via induction of cell apoptosis, oxidative stress, and inflammation; however, it's interfering with insulin signaling and decreased endothelial nitric oxide (NO) availability, resulting in the development of endothelial insulin resistance, which seems to be a major underlying mechanism for hyperuricemia-induced endothelial dysfunction. Here, we elaborate on how hyperuricemia induces endothelial insulin resistance through the disruption of insulin-stimulated endothelial NO synthesis. High UA concentrations decrease insulin-induced NO synthesis within the endothelial cells by interfering with insulin signaling at either the receptor or post-receptor levels (i.e., proximal and distal steps). At the proximal post-receptor level, UA impairs the function of the insulin receptor substrate (IRS) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) in the insulin signaling pathway. At the distal level, high UA concentrations impair endothelial NO synthase (eNOS)-NO system by decreasing eNOS expression and activity as well as by direct inactivation of NO. Clinically, UA-induced endothelial insulin resistance is translated into impaired endothelial function, impaired NO-dependent vasodilation, and the development of systemic insulin resistance. UA-lowering drugs may improve endothelial function in subjects with hyperuricemia.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA.,Graduate Program in Biology, City University of New York Graduate Center, New York, NY, 10016, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, P.O. Box: 19395-4763, VelenjakTehran, Iran.
| |
Collapse
|
25
|
Zhao Z, Zhao Y, Zhang Y, Shi W, Li X, Shyy JYJ, He M, Wang L. Gout-induced endothelial impairment: The role of SREBP2 transactivation of YAP. FASEB J 2021; 35:e21613. [PMID: 33977576 DOI: 10.1096/fj.202100337r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022]
Abstract
Gout is a multifaceted inflammatory disease involving vascular impairments induced by hyperuricemia. Experiments using human umbilical vein endothelial cells treated with uric acid (UA), monosodium urate (MSU), or serum from gout patients showed increased expression of pro-inflammatory genes (ie, VCAM1, ICAM1, CYR61, CCNA1, and E2F1) with attendant increase in monocyte adhesion. Mechanistically, UA- or MSU-induced SREBP2 expression and its transcriptional activity. RNA sequencing analysis and real-time PCR showed the induction of YAP signaling and pro-inflammatory pathways in HUVECs transfected with adenovirus-SREBP2. The SREBP2 knockdown by siRNA partially abolished UA- or MSU-induced YAP activity, pro-inflammatory gene expression, and monocytes adhesion. Vascular intima from transgenic mice overexpressing SREBP2 in endothelium or mice with hyperuricemia exhibited activated YAP signaling and increased expression of pro-inflammatory genes. Betulin, an SREBP pharmacological inhibitor, attenuated the UA-, MSU-, or gout serum-induced endothelial cell inflammation and dysfunction. In the human study, endothelial cell function, assessed by EndoPAT, was negatively correlated with serum UA level among gouty patients and healthy controls. Collectively, UA or MSU causes endothelial dysfunction via SREBP2 transactivation of YAP. Betulin inhibition of SREBP2 may restrain gout-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Zunlan Zhao
- Department of General Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yingshuai Zhao
- Department of General Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Zhang
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Weili Shi
- Department of General Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiqing Li
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ming He
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Liuyi Wang
- Department of General Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
26
|
Gohda T, Yanagisawa N, Murakoshi M, Ueda S, Nishizaki Y, Nojiri S, Ohashi Y, Ohno I, Shibagaki Y, Imai N, Iimuro S, Kuwabara M, Hayakawa H, Kimura K, Hosoya T, Suzuki Y. Association Between Kidney Function Decline and Baseline TNFR Levels or Change Ratio in TNFR by Febuxostat Chiefly in Non-diabetic CKD Patients With Asymptomatic Hyperuricemia. Front Med (Lausanne) 2021; 8:634932. [PMID: 34322499 PMCID: PMC8310915 DOI: 10.3389/fmed.2021.634932] [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] [Received: 11/29/2020] [Accepted: 06/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The levels of circulating tumor necrosis factor receptor (TNFR) 1 and 2 help predict the future decline of estimated glomerular filtration rate (eGFR) chiefly in patients with diabetes. It has been recently reported that the change ratio in TNFR1 by SGLT2 inhibitor treatment is also related with future GFR decline in patients with diabetes. The aims of this study are to investigate the association between baseline TNFR levels and early change in TNFR levels by the non-purine selective xanthine oxidase inhibitor, febuxostat, and future eGFR decline chiefly in chronic kidney disease (CKD) patients without diabetes. Methods: We conducted a post-hoc analysis of the FEATHER study on patients with asymptomatic hyperuricemia and CKD stage 3, who were randomly assigned febuxostat 40 mg/day or matched placebo. This analysis included 426 patients in whom baseline stored samples were available. Serum TNFR levels at baseline were measured using enzyme-linked immunosorbent assay. Those levels were also measured using 12-week stored samples from 197 randomly selected patients. Results: Compared with placebo, short-term febuxostat treatment significantly decreased the median percent change from baseline in serum uric acid (−45.05, 95% CI −48.90 to −41.24 mg/dL), TNFR1 (1.10, 95% CI−2.25 to 4.40), and TNFR2 (1.66, 95% CI −1.72 to 4.93), but not TNFR levels. Over a median follow-up of 105 weeks, 30 patients (7.0%) experienced 30% eGFR decline from baseline. In the Cox multivariate model, high levels of baseline TNFR predicted a 30% eGFR decline, even after adjusting for age, sex, systolic blood pressure, high sensitivity C-reactive protein, uric acid, and presence or absence of febuxostat treatment and diabetes, in addition to baseline albumin to creatinine ratio and eGFR. Conclusion: Early change in circulating TNFR levels failed to predict future eGFR decline; however, regardless of febuxostat treatment, the elevated baseline level of TNFR was a strong predictor of 30% eGFR decline even in chiefly non-diabetic CKD patients with asymptomatic hyperuricemia.
Collapse
Affiliation(s)
- Tomohito Gohda
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | | - Maki Murakoshi
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Seiji Ueda
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuji Nishizaki
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Shuko Nojiri
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Yasuo Ohashi
- Department of Integrated Science and Engineering for Sustainable Society, Chuo University, Tokyo, Japan
| | - Iwao Ohno
- Division of General Medicine, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naohiko Imai
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Satoshi Iimuro
- Innovation and Research Support Center, International University of Health and Welfare, Tokyo, Japan
| | - Masanari Kuwabara
- Intensive Care Unit, Department of Cardiology, Toranomon Hospital, Tokyo, Japan
| | | | | | - Tatsuo Hosoya
- Division of Chronic Kidney Disease Therapeutics, The Jikei University, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
The Entry and Egress of Monocytes in Atherosclerosis: A Biochemical and Biomechanical Driven Process. Cardiovasc Ther 2021; 2021:6642927. [PMID: 34345249 PMCID: PMC8282391 DOI: 10.1155/2021/6642927] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
In accordance with “the response to injury” theory, the entry of monocytes into the intima guided by inflammation signals, taking up cholesterol and transforming into foam cells, and egress from plaques determines the progression of atherosclerosis. Multiple cytokines and receptors have been reported to be involved in monocyte recruitment such as CCL2/CCR2, CCL5/CCR5, and CX3CL1/CX3CR1, and the egress of macrophages from the plaque like CCR7/CCL19/CCL21. Interestingly, some neural guidance molecules such as Netrin-1 and Semaphorin 3E have been demonstrated to show an inhibitory effect on monocyte migration. During the processes of monocytes recruitment and migration, factors affecting the biomechanical properties (e.g., the membrane fluidity, the deformability, and stiffness) of the monocytes, like cholesterol, amyloid-β peptide (Aβ), and lipopolysaccharides (LPS), as well as the biomechanical environment that the monocytes are exposed, like the extracellular matrix stiffness, mechanical stretch, blood flow, and hypertension, were discussed in the latter section. Till now, several small interfering RNAs (siRNAs), monoclonal antibodies, and antagonists for CCR2 have been designed and shown promising efficiency on atherosclerosis therapy. Seeking more possible biochemical factors that are chemotactic or can affect the biomechanical properties of monocytes, and uncovering the underlying mechanism, will be helpful in future studies.
Collapse
|
28
|
Mannarino MR, Pirro M, Gigante B, Savonen K, Kurl S, Giral P, Smit A, Veglia F, Tremoli E, Baldassarre D. Association Between Uric Acid, Carotid Intima-Media Thickness, and Cardiovascular Events: Prospective Results From the IMPROVE Study. J Am Heart Assoc 2021; 10:e020419. [PMID: 33998285 PMCID: PMC8483552 DOI: 10.1161/jaha.120.020419] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The association between elevated serum uric acid (SUA), cardiovascular disease (CVD) risk, and carotid atherosclerosis has long been explored, and contrasting results have been reported. Therefore, the role of SUA as an independent risk factor for vascular events (VEs) and carotid atherosclerosis deserves further attention. We investigated the relationship between SUA, incident VEs, carotid intima-media thickness (cIMT), and cIMT progression in subjects at moderate-to-high CVD risk. Methods and Results In the IMPROVE (IMT-Progression as Predictors of VEs) study, 3686 participants (median age 64 years; 48% men) with ≥ 3 vascular risk factors, free from VEs at baseline, were grouped according to SUA quartiles (division points: 244-284-328 µmol/L in women, 295-336-385 µmol/L in men). Carotid-IMT and its 15-month progression, along with incident VEs, were recorded. A U-shaped association between SUA and VEs was observed in men, with 2.4-fold (P = 0.004) and 2.5-fold (P = 0.002) increased CVD risk in the first and fourth SUA quartiles as compared with the second. Adjusted hazard ratios (HRs) for cerebro-VEs in men were the highest (first and fourth quartile versus second: HR, 5.3, P = 0.010 and HR, 4.4, P = 0.023, respectively). SUA level was independently associated with cIMT progression in men (β = 0.068, P = 0.014). No significant association between SUA levels, CVD end points, and cIMT progression were found in women. Conclusions Both low and high SUA levels are associated with an increased risk of VEs in men at moderate-to-high CVD risk but not in women. Only elevated SUA levels predict cIMT progression and at a lesser but not significant extent in women.
Collapse
Affiliation(s)
- Massimo R Mannarino
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases Department of Medicine and Surgery University of Perugia Perugia Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases Department of Medicine and Surgery University of Perugia Perugia Italy
| | - Bruna Gigante
- Unit of Cardiovascular Epidemiology Institute of Environmental Medicine Stockholm Sweden.,Division of Cardiovascular Medicine Department of Clinical Sciences Danderyd HospitalKarolinska Institutet Stockholm Sweden
| | - Kai Savonen
- Foundation for Research in Health Exercise and Nutrition Kuopio & Research Institute of Exercise Medicine Kuopio Finland.,Department of Clinical Physiology and Nuclear Medicine Kuopio University Hospital Kuopio Finland
| | - Sudhir Kurl
- Institute of Public Health and Clinical Nutrition University of Eastern Finland Kuopio Finland
| | - Philippe Giral
- Unités de Prévention Cardiovasculaire Service Endocrinologie-Metabolisme Assistance Publique - Hopitaux de ParisGroupe Hôpitalier Pitie-Salpetriere Paris France
| | - Andries Smit
- Department of Medicine University Medical Center Groningen Groningen the Netherlands.,Department of Medicine Isala Clinics Zwolle Zwolle the Netherlands
| | - Fabrizio Veglia
- Centro Cardiologico MonzinoIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Milan Italy
| | - Elena Tremoli
- Centro Cardiologico MonzinoIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Milan Italy
| | - Damiano Baldassarre
- Centro Cardiologico MonzinoIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Milan Italy.,Department of Medical Biotechnology and Translational Medicine Università degli Studi di Milano Milan Italy
| | | |
Collapse
|
29
|
Mozzini C, Girelli D, Setti A, Croce J, Stefanoni F, Castagna A, Pizzolo F, Friso S, Olivieri O, Martinelli N. Serum Uric Acid Levels, but Not rs7442295 Polymorphism of SCL2A9 Gene, Predict Mortality in Clinically Stable Coronary Artery Disease. Curr Probl Cardiol 2021; 46:100798. [PMID: 33540324 DOI: 10.1016/j.cpcardiol.2021.100798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 10/22/2022]
Abstract
Serum uric acid (SUA) has been associated with cardiovascular disease, but up to now whether SUA is an independent cardiovascular risk factor or merely a disease-related epiphenomenon remains still controversial. within the framework of the Verona Heart Study, we prospectively followed 703 subjects with angiographically demonstrated and clinically stable coronary artery disease between May 1996 and March 2007. At baseline, SUA levels were measured in all the patients. Genotype data of SCL2A9 rs7442295 polymorphism, which has been associated with SUA by genome-wide association studies, were available for 686 subjects (97.6%). After a median follow-up of 57 months, 116 patients (16.5%) had died, 83 (11.8%) because of cardiovascular causes. Patients with hyperuricemia, defined by SUA levels above the 75th percentile (≥0.41 mmol/L), had an increased total and cardiovascular mortality rate than those with SUA below this threshold level (23.3% vs 14.1%, P = 0.048 and 19.4% vs 9.2%, P = 0.001, respectively, by Kaplan-Meier with Log-Rank test). These associations were confirmed by Cox regression after adjustment for sex, age, other predictors of mortality, coronary revascularization, and drug therapies at discharge (hazard ratio for total mortality 1.87 [1.05-3.34], P = 0.033; hazard ratio for cardiovascular mortality 2.09 [1.03-4.25], P = 0.041). Although associated with SUA levels, rs7442295 polymorphism did not predict total or cardiovascular mortality. our data support that SUA may be a prognostic cardiovascular biomarker, predicting total and cardiovascular mortality in the setting of secondary prevention of coronary artery disease. On the other hand, SCL2A9 gene polymorphism, notwithstanding a clear influence on SUA levels, was not associated with mortality.
Collapse
Affiliation(s)
- Chiara Mozzini
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy.
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Angela Setti
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Jacopo Croce
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Filippo Stefanoni
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Francesca Pizzolo
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Simonetta Friso
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Oliviero Olivieri
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Nicola Martinelli
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| |
Collapse
|
30
|
Abstract
ABSTRACT Secondary brain injury following hemorrhagic shock (HS) is a frequent complication in patients, even in the absence of direct brain trauma, leading to behavioral changes and more specifically anxiety and depression. Despite preclinical studies showing inflammation and apoptosis in the brain after HS, none have addressed the impact of circulating mediators. Our group demonstrated an increased uric acid (UA) circulation in rats following HS. Since UA is implicated in endothelial dysfunction and inflammatory response, we hypothesized UA could alter the blood-brain barrier (BBB) and impact the brain. Male Wistar rats were randomly assigned to: SHAM, HS (hemorrhagic shock) and HS + U (hemorrhagic shock + 1.5 mg/kg of uricase). The uricase intervention, specifically targeting UA, was administered during fluid resuscitation. It prevented BBB dysfunction (fluorescein sodium salt permeability and expression of intercellular adhesion molecule-1) following HS. As for neuroinflammation, all of the results obtained (MPO activity; Iba1 and GFAP expression) showed a significant increase after HS, also prevented by the uricase. The same pattern was observed after quantification of apoptosis (caspase-3 activity and TUNEL) and neurodegeneration (Fluoro-Jade). Finally, the forced swim, elevated plus maze, and social interaction tests detected anxiety-like behavior after HS, which was blunted in rats treated with the uricase. In conclusion, we have identified UA as a new circulatory inflammatory mediator, responsible for brain alterations and anxious behavior after HS in a murine model. The ability to target UA holds the potential of an adjunctive therapeutic solution to reduce brain dysfunction related to hemorrhagic shock in human.
Collapse
|
31
|
Morris G, Puri BK, Olive L, Carvalho A, Berk M, Walder K, Gustad LT, Maes M. Endothelial dysfunction in neuroprogressive disorders-causes and suggested treatments. BMC Med 2020; 18:305. [PMID: 33070778 PMCID: PMC7570030 DOI: 10.1186/s12916-020-01749-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/16/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Potential routes whereby systemic inflammation, oxidative stress and mitochondrial dysfunction may drive the development of endothelial dysfunction and atherosclerosis, even in an environment of low cholesterol, are examined. MAIN TEXT Key molecular players involved in the regulation of endothelial cell function are described, including PECAM-1, VE-cadherin, VEGFRs, SFK, Rho GEF TRIO, RAC-1, ITAM, SHP-2, MAPK/ERK, STAT-3, NF-κB, PI3K/AKT, eNOS, nitric oxide, miRNAs, KLF-4 and KLF-2. The key roles of platelet activation, xanthene oxidase and myeloperoxidase in the genesis of endothelial cell dysfunction and activation are detailed. The following roles of circulating reactive oxygen species (ROS), reactive nitrogen species and pro-inflammatory cytokines in the development of endothelial cell dysfunction are then described: paracrine signalling by circulating hydrogen peroxide, inhibition of eNOS and increased levels of mitochondrial ROS, including compromised mitochondrial dynamics, loss of calcium ion homeostasis and inactivation of SIRT-1-mediated signalling pathways. Next, loss of cellular redox homeostasis is considered, including further aspects of the roles of hydrogen peroxide signalling, the pathological consequences of elevated NF-κB, compromised S-nitrosylation and the development of hypernitrosylation and increased transcription of atherogenic miRNAs. These molecular aspects are then applied to neuroprogressive disorders by considering the following potential generators of endothelial dysfunction and activation in major depressive disorder, bipolar disorder and schizophrenia: NF-κB; platelet activation; atherogenic miRs; myeloperoxidase; xanthene oxidase and uric acid; and inflammation, oxidative stress, nitrosative stress and mitochondrial dysfunction. CONCLUSIONS Finally, on the basis of the above molecular mechanisms, details are given of potential treatment options for mitigating endothelial cell dysfunction and activation in neuroprogressive disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | | | - Lisa Olive
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- School of Psychology, Faculty of Health, Deakin University, Geelong, Australia
| | - Andre Carvalho
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia.
- Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Ken Walder
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Lise Tuset Gustad
- Department of Circulation and medical imaging, Norwegian University of Technology and Science (NTNU), Trondheim, Norway
- Nord-Trøndelag Hospital Trust, Levanger Hospital, Levanger, Norway
| | - Michael Maes
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
32
|
Chen MQ, Wang HY, Shi WR, Sun YX. Estimate of prevalent hyperuricemia by systemic inflammation response index: results from a rural Chinese population. Postgrad Med 2020; 133:242-249. [PMID: 32921215 DOI: 10.1080/00325481.2020.1809870] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Hyperuricemia is a common metabolic disease that is intimately correlated with inflammation. Our study aimed to investigate the value of systemic inflammation response index as a novel inflammatory marker to estimate hyperuricemia in the rural Chinese population. METHODS This cross-sectional study used the data of 8,095 Chinese men and women aged ≥35 years from the 2012-2013 Northeast China Rural Cardiovascular Health Study. RESULTS The overall prevalence of hyperuricemia was 12.84%. After fully adjusting for potential confounders, each SD increase of SIRI in men and women caused a 21.4% and 37.0% additional risk, respectively, for hyperuricemia. Moreover, smooth curve fitting and subgroup analyses corroborated the linearity and robustness of this correlation. ROC analysis showed the ability of SIRI to estimate hyperuricemia was significantly improved in females (0.741 vs 0.745, P = 0.043), but not in males (0.710 vs 0.714, P = 0.105). The net reclassification improvement (NRI, 0.120 in men vs 0.166 in women) and integrated discrimination improvement (IDI, 0.002 in men vs 0.006 in women) showed a significant improvement for both genders. CONCLUSIONS Our present study suggests a linear and robust relationship between SIRI and prevalent hyperuricemia, which implicates the value of SIRI to optimize the risk stratification and prevention of hyperuricemia.
Collapse
Affiliation(s)
- Meng-Qi Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Hao-Yu Wang
- Department of Cardiology, Coronary Heart Disease Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wen-Rui Shi
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Ying-Xian Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
33
|
Fonseca W, Malinczak CA, Schuler CF, Best SK, Rasky AJ, Morris SB, Cui TX, Popova AP, Lukacs NW. Uric acid pathway activation during respiratory virus infection promotes Th2 immune response via innate cytokine production and ILC2 accumulation. Mucosal Immunol 2020; 13:691-701. [PMID: 32047272 PMCID: PMC7316593 DOI: 10.1038/s41385-020-0264-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/05/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
Abstract
Respiratory syncytial virus (RSV) infects a majority of infants and can cause severe disease leading to increased risk to develop asthma later in life. In the present studies we detected high levels of uric acid pathway components during RSV infection and examined whether they altered the pathogenesis of RSV infection. Inhibition of uric acid (UA) pathway activation during RSV infection in airway epithelial cells using XOI decreased the expression of IL-33, thymic stromal lymphopoietin (TSLP), and CCL2. In addition, treatment of RSV infected bone marrow-derived macrophages with XOI decreased production of IL-1β. Thus, UA activation of different cell populations contributes different innate immune mediators that promote immunopathogenesis. When mice were treated with XOI or interleukin-1 receptor antagonist (IL1-ra) during RSV infection decreased pulmonary mucus was observed along with significantly reduced numbers of ILC2 and macrophages, accompanied by decreased IL-33 in bronchoalveolar lavage of the treated mice. These findings provide mechanistic insight into the development of RSV immunopathology and indicate that xanthine metabolites and UA are key immunoregulator molecules during RSV infection. Moreover, these findings suggest uric acid and IL-1β as possible therapeutic targets to attenuate severe RSV disease.
Collapse
Affiliation(s)
- Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA 48109
| | | | - Charles F. Schuler
- Department of Internal Medicine, Division of Allergy and Clinical Immunology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Shannon K.K. Best
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Susan B Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Tracy X. Cui
- Division of Pediatric Pulmonology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Antonia P. Popova
- Division of Pediatric Pulmonology, University of Michigan, Ann Arbor, MI, USA 48109
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA. .,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
34
|
Ma Q, Honarpisheh M, Li C, Sellmayr M, Lindenmeyer M, Böhland C, Romagnani P, Anders HJ, Steiger S. Soluble Uric Acid Is an Intrinsic Negative Regulator of Monocyte Activation in Monosodium Urate Crystal-Induced Tissue Inflammation. THE JOURNAL OF IMMUNOLOGY 2020; 205:789-800. [PMID: 32561569 DOI: 10.4049/jimmunol.2000319] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
Although monosodium urate (MSU) crystals are known to trigger inflammation, published data on soluble uric acid (sUA) in this context are discrepant. We hypothesized that diverse sUA preparation methods account for this discrepancy and that an animal model with clinically relevant levels of asymptomatic hyperuricemia and gouty arthritis can ultimately clarify this issue. To test this, we cultured human monocytes with different sUA preparation solutions and found that solubilizing uric acid (UA) by prewarming created erroneous results because of UA microcrystal contaminants triggering IL-1β release. Solubilizing UA with NaOH avoided this artifact, and this microcrystal-free preparation suppressed LPS- or MSU crystal-induced monocyte activation, a process depending on the intracellular uptake of sUA via the urate transporter SLC2A9/GLUT9. CD14+ monocytes isolated from hyperuricemic patients were less responsive to inflammatory stimuli compared with monocytes from healthy individuals. Treatment with plasma from hyperuricemic patients impaired the inflammatory function of CD14+ monocytes, an effect fully reversible by removing sUA from hyperuricemic plasma. Moreover, Alb-creERT2;Glut9 lox/lox mice with hyperuricemia (serum UA of 9-11 mg/dl) showed a suppressed inflammatory response to MSU crystals compared with Glut9 lox/lox controls without hyperuricemia. Taken together, we unravel a technical explanation for discrepancies in the published literature on immune effects of sUA and identify hyperuricemia as an intrinsic suppressor of innate immunity, in which sUA modulates the capacity of monocytes to respond to danger signals. Thus, sUA is not only a substrate for the formation of MSU crystals but also an intrinsic inhibitor of MSU crystal-induced tissue inflammation.
Collapse
Affiliation(s)
- Qiuyue Ma
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Mohsen Honarpisheh
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Chenyu Li
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Markus Sellmayr
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Maja Lindenmeyer
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Claudia Böhland
- Department of Radiation Oncology, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Germany; and
| | - Paola Romagnani
- Department of Biomedical Experimental and Clinical Sciences "Maria Serio," University of Florence, 50139 Florence, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig Maximilian University of Munich, 80336 Munich, Bavaria, Germany;
| |
Collapse
|
35
|
Abstract
: Uric acid levels are higher in humans than in other mammals. Best known as an extracellular antioxidant, uric acid also increases salt sensitivity, fat storage, and lipogenesis. Xanthine oxidase-related oxidative stress may also induce endothelial dysfunction and renal vasoconstriction. Renal structure abnormalities contribute to salt-sensitive and uric acid-independent hypertension. Maternal hyperuricemia during pregnancy and hyperuricemia early in life are likewise independent risk factors for hypertension. Genetic polymorphism is potentially involved in the activity of xanthine oxidoreductase, but further studies are needed. Xanthine oxidase inhibition consistently decreases blood pressure in younger hypertensive patients, albeit modestly. Hyperuricemia affects one out of five adults as a result of the Western diet, insulin resistance, and renal dysfunction. This review advocates lifestyle changes to maintain uric acid levels within the normal range in young (pre)hypertensive individuals or normotensives with a family history of hypertension, metabolic disorders, or obesity; moreover, antihypertensive medications that increase uric acid levels should be avoided.
Collapse
|
36
|
Wang G, Zuo T, Li R. The mechanism of Arhalofenate in alleviating hyperuricemia―Activating
PPARγ
thereby reducing caspase‐1 activity. Drug Dev Res 2020; 81:859-866. [PMID: 32506648 DOI: 10.1002/ddr.21699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Guihong Wang
- Department of Rheumatology and Immunology Anqing Hospital, affiliated Hospital of Anhui Medical University Anqing China
| | - Ting Zuo
- Department of Rheumatology and Immunology Anqing Hospital, affiliated Hospital of Anhui Medical University Anqing China
| | - Ran Li
- Department of Rheumatology and Immunology Anqing Hospital, affiliated Hospital of Anhui Medical University Anqing China
| |
Collapse
|
37
|
Association between serum copper levels and prevalence of hyperuricemia: a cross-sectional study. Sci Rep 2020; 10:8687. [PMID: 32457333 PMCID: PMC7250918 DOI: 10.1038/s41598-020-65639-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/07/2020] [Indexed: 12/28/2022] Open
Abstract
Hyperuricemia has been recognized as a worldwide public health concern. This study was conducted to examine the association between serum copper (Cu) concentration and the prevalence of hyperuricemia in a middle-aged and elderly population. Serum Cu concentration was measured by Roche modular P800 using the PAESA method. Serum uric acid (UA) concentration was detected by a Beckman Coulter AU 5800. Presence of hyperuricemia was defined as serum UA ≥ 416 μmol/L for men and ≥360 μmol/L for women. The association between serum Cu concentration and the prevalence of hyperuricemia was evaluated by logistic regression. The prevalence of hyperuricemia was 17.6% (n = 6,212) in the present study. Relative to the lowest quintile, the age- and sex-adjusted odds ratios for hyperuricemia were 1.38 (95% CI: 1.12 to 1.70), 1.34 (95% CI: 1.07 to 1.66), and 1.53 (95% CI: 1.23 to 1.91) in the third, fourth, and fifth serum Cu concentration quintiles (P for trend < 0.001). Similar results were found both in men and women subgroups. None of the findings were materially altered after adjustment for additional potential confounders. In conclusion, in this population-based cross-sectional study, serum Cu concentration was positively associated with the prevalence of hyperuricemia.
Collapse
|
38
|
Jung SW, Kim SM, Kim YG, Lee SH, Moon JY. Uric acid and inflammation in kidney disease. Am J Physiol Renal Physiol 2020; 318:F1327-F1340. [PMID: 32223310 DOI: 10.1152/ajprenal.00272.2019] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Asymptomatic hyperuricemia is frequently observed in patients with kidney disease. Although a substantial number of epidemiologic studies have suggested that an elevated uric acid level plays a causative role in the development and progression of kidney disease, whether hyperuricemia is simply a result of decreased renal excretion of uric acid or is a contributor to kidney disease remains a matter of debate. Over the last two decades, multiple experimental studies have expanded the knowledge of the biological effects of uric acid beyond its role in gout. In particular, uric acid induces immune system activation and alters the characteristics of resident kidney cells, such as tubular epithelial cells, endothelial cells, and vascular smooth muscle cells, toward a proinflammatory and profibrotic state. These findings have led to an increased awareness of uric acid as a potential and modifiable risk factor in kidney disease. Here, we discuss the effects of uric acid on the immune system and subsequently review the effects of uric acid on the kidneys mainly in the context of inflammation.
Collapse
Affiliation(s)
- Su Woong Jung
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| | - Su-Mi Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| | - Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| | - Sang-Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
39
|
Hydralazine improves ischemia-induced neovasculogenesis via xanthine-oxidase inhibition in chronic renal insufficiency. Pharmacol Res 2019; 151:104509. [PMID: 31678640 DOI: 10.1016/j.phrs.2019.104509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/13/2019] [Accepted: 10/21/2019] [Indexed: 01/10/2023]
Abstract
Oxidative stress is related to the progression of renal diseases and modulation of oxidative stress can lead to a reduction in vascular events in patients with chronic renal insufficiency (CRI). Indoxyl sulfate (IS) and xanthine oxidase (XO) are related to impaired neovasculogenesis in CRI. Hydralazine is suggested for blood pressure control in CRI. This study aimed to investigate whether hydralazine could improve ischemia-induced neovasculogenesis in CRI animals by reducing reactive oxygen species (ROS) levels. Mice underwent subtotal nephrectomy or sham surgery. Nitrendipine, probenecid, and allopurinol were used to reduce blood pressure, uric acid (UA), and XO activity levels, respectively, for comparison. Blood pressure, XO activity and UA levels that were increased after subtotal nephrectomy were reduced by hydralazine treatment. Allopurinol decreased blood XO activity and UA levels. Only hydralazine and allopurinol increased the number of circulating endothelial progenitor cells (EPCs) and improved neovasculogenesis in CRI mice. IS activated XO mRNA and ROS and inhibited the functions of EPCs and endothelial cells, which could be reversed by hydralazine. However, no additional beneficial effects were observed when XO was inhibited with both hydralazine and siRNA. In conclusion, hydralazine, as a potential XO inhibitor, not only reduced blood pressure and UA levels but also increased the number of circulating EPCs and improved neovasculogenesis in CRI animals. Hydralazine directly inhibited IS-induced ROS and XO activation in EPCs and endothelial cells, and restored their functions in vitro. Future studies should evaluate whether hydralazine could provide additional vascular protection in patients with CRI.
Collapse
|
40
|
Zhang JX, Lin X, Xu J, Tang F. Hyperuricemia Inhibition Protects SD Rats Against Fructose-Induced Obesity Hypertension Via Modulation of Inflammation and Renin-Angiotensin System in Adipose Tissue. Exp Clin Endocrinol Diabetes 2019; 129:314-321. [PMID: 31683330 DOI: 10.1055/a-1023-6710] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The present study was aimed to reveal the relationship between uric acid and fructose-induced obesity hypertension and its mechanisms. METHODS A rat model with obesity hypertension was induced by a high-fructose diet. In the experiment I, the rats were fed with fructose for 8 wks along with allopurinol or benzbromarone at the beginning. In the experiment II, the rats were fed with fructose for 8 wks firstly. And then, these rats were treated with allopurinol or benzbromarone for additional 6 wks. RESULTS Fructose-fed rats showed hyperuricemia, abdominal obesity hypertension and an activation in adipose renin-angiotensin system (RAS). Also, fructose-fed rats had higher levels of proinflammatory cytokines and more macrophages infiltrating in adipose tissue. In the experiment I, allopurinol and benzbromarone significantly reduced serum uric acid at 8 wk. Adipose RAS overactivation, adipose inflammatory responses and the development of obesity hypertension were all effectively prevented by hyperuricemia inhibition. In the experiment II, 6-wk treatment with allopurinol and benzbromarone significantly decreased serum uric acid, downregulated adipose RAS, abolished adipose inflammation and improved obesity hypertension. CONCLUSION In conclusion, urate-lowering therapy protects rats against fructose-induced obesity hypertension. The mechanisms appear to be via downregulated adipose RAS and reduced inflammation in adipose tissue.
Collapse
Affiliation(s)
- Jun Xia Zhang
- Department of Endocrinology, Central Theater Command General Hospital of the Chinese PLA, Wuhan, China
| | - Xue Lin
- Department of Endocrinology, Central Theater Command General Hospital of the Chinese PLA, Wuhan, China
| | - Jinxiu Xu
- Department of Endocrinology, Central Theater Command General Hospital of the Chinese PLA, Wuhan, China
| | - Feng Tang
- Department of Endocrinology, Central Theater Command General Hospital of the Chinese PLA, Wuhan, China
| |
Collapse
|
41
|
New Insights into the Roles of Monocytes/Macrophages in Cardiovascular Calcification Associated with Chronic Kidney Disease. Toxins (Basel) 2019; 11:toxins11090529. [PMID: 31547340 PMCID: PMC6784181 DOI: 10.3390/toxins11090529] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is an important cause of death in patients with chronic kidney disease (CKD), and cardiovascular calcification (CVC) is one of the strongest predictors of CVD in this population. Cardiovascular calcification results from complex cellular interactions involving the endothelium, vascular/valvular cells (i.e., vascular smooth muscle cells, valvular interstitial cells and resident fibroblasts), and monocyte-derived macrophages. Indeed, the production of pro-inflammatory cytokines and oxidative stress by monocyte-derived macrophages is responsible for the osteogenic transformation and mineralization of vascular/valvular cells. However, monocytes/macrophages show the ability to modify their phenotype, and consequently their functions, when facing environmental modifications. This plasticity complicates efforts to understand the pathogenesis of CVC-particularly in a CKD setting, where both uraemic toxins and CKD treatment may affect monocyte/macrophage functions and thereby influence CVC. Here, we review (i) the mechanisms by which each monocyte/macrophage subset either promotes or prevents CVC, and (ii) how both uraemic toxins and CKD therapies might affect these monocyte/macrophage functions.
Collapse
|
42
|
Yin W, Zhou QL, OuYang SX, Chen Y, Gong YT, Liang YM. Uric acid regulates NLRP3/IL-1β signaling pathway and further induces vascular endothelial cells injury in early CKD through ROS activation and K + efflux. BMC Nephrol 2019; 20:319. [PMID: 31412804 PMCID: PMC6694569 DOI: 10.1186/s12882-019-1506-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) has been considered as a major health problem in the world. Increasing uric acid (UA) could induce vascular endothelial injury, which is closely related to microinflammation, oxidative stress, and disorders of lipids metabolism. However, the specific mechanism that UA induces vascular endothelial cells injury in early CKD remains unknown. METHODS Human umbilical vein endothelial cells (HUVECs) were cultured and subjected to different concentrations of UA for different periods. Early CKD rat model with elevated serum UA was established. Western blotting and quantitative real-time PCR (qPCR) were applied for measuring protein and mRNA expression of different cytokines. The animals were sacrificed and blood samples were collected for measurement of creatinine, UA, IL-1β, TNF-α, and ICAM-1. Renal tissues were pathologically examined by periodic acid-Schiff (PAS) or hematoxylin-eosin (HE) staining. RESULTS The expression of IL-1β, ICAM-1, NLRP3 complexes, and activation of NLRP3 inflammasome could be induced by UA, but the changes induced by UA were partially reversed by siRNA NLRP3 or caspase 1 inhibitor. Furthermore, we identified that UA regulated the activation of NLRP3 inflammasome by activating ROS and K+ efflux. In vivo results showed that UA caused the vascular endothelial injury by activating NLRP3/IL-1β pathway. While allopurinol could reduce UA level and may have protective effects on cardiovascular system. CONCLUSIONS UA could regulate NLRP3/IL-1β signaling pathway through ROS activation and K+ efflux and further induce vascular endothelial cells injury in early stages of CKD.
Collapse
Affiliation(s)
- Wei Yin
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China
| | - Qiao-Ling Zhou
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Sha-Xi OuYang
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China
| | - Ying Chen
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China
| | - Yu-Ting Gong
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China
| | - Yu-Mei Liang
- Department of Nephrology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, No.61, West Jiefang Road, Changsha, 410005, Hunan Province, People's Republic of China.
| |
Collapse
|
43
|
Zhang B, Duan M, Long B, Zhang B, Wang D, Zhang Y, Chen J, Huang X, Jiao Y, Zhu L, Zeng X. Urate transport capacity of glucose transporter 9 and urate transporter 1 in cartilage chondrocytes. Mol Med Rep 2019; 20:1645-1654. [PMID: 31257523 PMCID: PMC6625399 DOI: 10.3892/mmr.2019.10426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 05/31/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic gouty arthritis, caused by a persistent increase in, and the deposition of, soluble uric acid (sUA), can induce pathological chondrocyte destruction; however, the effects of urate transport and intracellular sUA on chondrocyte functionality and viability are yet to be fully determined. Thus, the aim of the present study was to investigate the presence and functionality of a urate transport system in chondrocytes. The expression profiles of two primary urate reabsorptive transporters, glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1), in human articular cartilage and chondrocyte cell lines were examined via western blotting, reverse transcription-quantitative PCR, immunohistochemistry and immunofluorescence. Then, chondrocytes were incubated with exogenous sUA at increasing concentrations. Negative control assays were conducted via the specific knockdown of GLUT9 and URAT1 with lentiviral short hairpin (sh)RNAs, and by pretreatment with benzbromarone, a known inhibitor of the two transporters. Intracellular UA concentrations were measured using colorimetric assays. The expression levels of GLUT9 and URAT1 were determined in cartilage tissues and chondrocyte cell lines. Incubation of chondrocytes with sUA led to a concentration-dependent increase in intracellular urate concentrations, which was inhibited by GLUT9 or URAT1 knockdown, or by benzbromarone pretreatment (27.13±2.70, 44.22±2.34 and 58.46±2.32% reduction, respectively). In particular, benzbromarone further decreased the already-reduced intracellular UA concentrations in HC-shGLUT9 and HC-shURAT1 cells by 46.79±2.46 and 39.79±2.22%, respectively. Cells overexpressing GLUT9 and URAT1 were used as the positive cell control, which showed increased intracellular UA concentrations that could be reversed by treatment with benzbromarone. In conclusion, chondrocytes may possess an active UA transport system. GLUT9 and URAT1 functioned synergistically to transport UA into the chondrocyte cytoplasm, which was inhibited by specific gene knockdowns and drug-induced inhibition. These results may be fundamental in the further investigation of the pathological changes to chondrocytes induced by sUA during gouty arthritis, and identified UA transport processes as potential targets for the early control of chronic gouty arthritis.
Collapse
Affiliation(s)
- Bingqing Zhang
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Mengyuan Duan
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Bo Long
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Baozhong Zhang
- Department of Orthopedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Dongmei Wang
- Department of Neurology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong 510515, P.R. China
| | - Yun Zhang
- Department of General Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Jialin Chen
- Department of General Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xiaoming Huang
- Department of General Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yang Jiao
- Department of General Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Lei Zhu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Xuejun Zeng
- Department of General Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
44
|
Nigam SK, Bush KT. Uraemic syndrome of chronic kidney disease: altered remote sensing and signalling. Nat Rev Nephrol 2019; 15:301-316. [PMID: 30728454 PMCID: PMC6619437 DOI: 10.1038/s41581-019-0111-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Uraemic syndrome (also known as uremic syndrome) in patients with advanced chronic kidney disease involves the accumulation in plasma of small-molecule uraemic solutes and uraemic toxins (also known as uremic toxins), dysfunction of multiple organs and dysbiosis of the gut microbiota. As such, uraemic syndrome can be viewed as a disease of perturbed inter-organ and inter-organism (host-microbiota) communication. Multiple biological pathways are affected, including those controlled by solute carrier (SLC) and ATP-binding cassette (ABC) transporters and drug-metabolizing enzymes, many of which are also involved in drug absorption, distribution, metabolism and elimination (ADME). The remote sensing and signalling hypothesis identifies SLC and ABC transporter-mediated communication between organs and/or between the host and gut microbiota as key to the homeostasis of metabolites, antioxidants, signalling molecules, microbiota-derived products and dietary components in body tissues and fluid compartments. Thus, this hypothesis provides a useful perspective on the pathobiology of uraemic syndrome. Pathways considered central to drug ADME might be particularly important for the body's attempts to restore homeostasis, including the correction of disturbances due to kidney injury and the accumulation of uraemic solutes and toxins. This Review discusses how the remote sensing and signalling hypothesis helps to provide a systems-level understanding of aspects of uraemia that could lead to novel approaches to its treatment.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Kevin T Bush
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
45
|
Zhou Y, Zhao M, Pu Z, Xu G, Li X. Relationship between oxidative stress and inflammation in hyperuricemia: Analysis based on asymptomatic young patients with primary hyperuricemia. Medicine (Baltimore) 2018; 97:e13108. [PMID: 30544373 PMCID: PMC6310523 DOI: 10.1097/md.0000000000013108] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The average age of hyperuricemia patients has gradually decreased, but young patients with primary hyperuricemia often do not exhibit clinical symptoms and have not received sufficient attention. However, a lack of symptoms with primary hyperuricemia does not mean that high serum uric acid (UA) levels cannot lead to pathological effects, such as oxidative stress and inflammation, and the specific damage is still unclear. We aimed to determine the relationship between oxidative stress and inflammation to explore the possible role of pathological damage in asymptomatic young patients with primary hyperuricemia.A total of 333 participants were enrolled in our study: 158 asymptomatic young patients with primary hyperuricemia and 175 healthy persons from a health check-up population. Malondialdehyde (MDA), superoxide dismutase (SOD), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and general biochemical markers were measured.We found no differences in biochemical markers (fasting glucose, TG, TC, LDL-C, HDL-C, SCr, BUN, AST, and ALT) between the patients and healthy persons. Subsequent analyses of oxidative stress and inflammation revealed that the serum levels of MDA, IL-6, and TNF-α in the patients were significantly higher than those in the healthy control group (P < .001), and the SOD activity was significantly lower (P < .001). As the UA levels increased, MDA increased significantly and SOD decreased significantly; likewise, IL-6 and TNF-α increased significantly as the UA level increased. MDA showed a significant positive correlation with IL-6 (r = 0.367, P < .001) and TNF-α (r = 0.319, P < .001), and SOD was negatively correlated with IL-6 (r = -0.241, P < .01) and TNF-α (r = -0.308, P < .001). Multivariable logistic regression analysis showed that UA (OR: 2.379, 95% CI: 1.698-3.286, P < .001; OR: 3.261, 95% CI: 1.729-3.857, P < .001; for IL-6 and TNF-α, respectively) and MDA (OR: 1.836, 95% CI: 1.283-2.517, P < .01; OR: 2.532, 95% CI: 1.693-3.102, P < .001; for IL-6 and TNF-α, respectively) were risk factors for high IL-6 and TNF-α and that SOD (OR: 0.517, 95% CI: 0.428-0.763, P < .01; OR: 0.603, 95% CI: 0.415-0.699, P < .001; for IL-6 and TNF-α, respectively) was a protective factor.In our study, some abnormal pathological effects were found in asymptomatic young patients with hyperuricemia, suggesting that in young hyperuricemia patients, oxidative stress, inflammation and the inflammatory response may be related to the oxidative stress induced by UA. Therefore, we should pay more attention to the pathological damage caused by these alterations.
Collapse
|
46
|
Fu X, Niu N, Li G, Xu M, Lou Y, Mei J, Liu Q, Sui Z, Sun J, Qu P. Blockage of macrophage migration inhibitory factor (MIF) suppressed uric acid-induced vascular inflammation, smooth muscle cell de-differentiation, and remodeling. Biochem Biophys Res Commun 2018; 508:440-444. [PMID: 30502082 DOI: 10.1016/j.bbrc.2018.10.093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/15/2018] [Indexed: 11/16/2022]
Abstract
Hyperuricemia contributes to vascular injury and dysfunction, yet the potential mechanisms are not well understood. Uric acid (UA) has been found to stimulate macrophage migration inhibitory factor (MIF) up-regulation in renal tubules from rats subjected to UA-induced nephropathy. Given that MIF is able to induce vascular smooth muscle cell (VSMC) de-differentiation (from contractile state to a secretory state), we thus hypothesized that UA-induced vascular injury is via up-regulating of MIF in VSMCs, which enhancing vascular inflammation and VSMC transition. Within a mouse model of UA injection (500 mg/kg, twice/day, 14 days), we measured circulating and vascular MIF levels under UA stimulation at 6 h, day 1, and 14. We tested the efficacy of MIF inhibitor (10 mg/kg, twice/day, 14 days) on UA-induced vascular inflammation and remodeling. High plasma level of UA induced vascular MIF release into the plasma at acute phase. In the chronic phase, the protein level of MIF is up-regulated in the vessels. MIF inhibitor suppressed vascular inflammatory responses, repressed VSMC de-differentiation, and attenuated vascular remodeling and dysfunction following UA stimulation. Knockdown of MIF in cultured VSMCs repressed UA-induced de-differentiation. Our results provided a novel mechanism for MIF-mediated vascular injury in response to UA stimulation, and suggested that anti-MIF interventions may be of therapeutic value in hyperuricemic patients.
Collapse
Affiliation(s)
- Xiaodan Fu
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Nan Niu
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Guihua Li
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Mingxi Xu
- Department of Rheumatology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yu Lou
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Jiajie Mei
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Qizhi Liu
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Zheng Sui
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Jingyi Sun
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Peng Qu
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
47
|
Yang H, Bai W, Gao L, Jiang J, Tang Y, Niu Y, Lin H, Li L. Mangiferin alleviates hypertension induced by hyperuricemia via increasing nitric oxide releases. J Pharmacol Sci 2018; 137:154-161. [PMID: 29934052 DOI: 10.1016/j.jphs.2018.05.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 05/06/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022] Open
Abstract
Mangiferin, a natural glucosyl xanthone, was confirmed to be an effective uric acid (UA)- lowering agent with dual action of inhibiting production and promoting excretion of UA. In this study, we aimed to evaluate the effect of mangiferin on alleviating hypertension induced by hyperuricemia. Mangiferin (30, 60, 120 mg/kg) was administered intragastrically to hyperuricemic rats induced by gavage with potassium oxonate (750 mg/kg). Systolic blood pressure (SBP), serum levels of UA, nitric oxide (NO), C-reactionprotein (CRP) and ONOO- were measured. The mRNA and protein levels of endothelial nitric oxide synthase (eNOS), intercellular adhesion molecule-1 (ICAM-1), CRP were also analyzed. Human umbilical vein endothelial cells (HUVECs) were used in vitro studies. Administration of mangiferin significantly decreased the serum urate level and SBP at 8 weeks and last to 12 weeks. Further more, mangiferin could increase the release of NO and decrease the level of CRP in blood. In addition, mangiferin reversed the protein expression of eNOS, CRP, ICAM-1 and ONOO- in aortic segments in hyperuricemic rats. The results in vitro were consistent with the observed results in vivo. Taken together, these data suggested that mangiferin has played an important part in alleviating hypertension induced by hyperuricemia via increasing NO secretion and improving endothelial function.
Collapse
Affiliation(s)
- Hua Yang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenwei Bai
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihui Gao
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, China
| | - Jun Jiang
- The Third People's Hospital of Yunnan Province, Kunming, China
| | | | - Yanfen Niu
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, China
| | - Hua Lin
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, China
| | - Ling Li
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, China.
| |
Collapse
|
48
|
Li Y, Fan X, Li C, Zhi X, Peng L, Han H, Sun B. The relationships among hyperuricemia, body mass index and impaired renal function in type 2 diabetic patients. Endocr J 2018; 65:281-290. [PMID: 29237999 DOI: 10.1507/endocrj.ej17-0266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Chronic kidney disease (CKD) is a common chronic microvascular complication and the major cause of death in diabetic patients. This study was conceived to explore the possible mechanisms of how hyperuricemia and obesity contribute to renal function impairment in type 2 diabetic (T2DM) patients. A cross-sectional study in 609 participants recruited from a T2DM population in North China was conducted. The multiplicative interaction between body mass index (BMI) and uric acid (UA) level was assessed using an interaction term in a logistic regression analysis. Our results indicate that male T2DM patients having higher BMI (OR 1.711, p = 0.038), blood urine nitrogen (BUN) (OR 1.100, p = 0.034), and 24-hour urinary micro-albumin levels (OR 1.004, p = 0.021) were much more likely to have high UA. Whereas, for female T2DM patients, the OR of BMI, BUN, and triglyceride were 1.169 (p = 0.001), 1.337 (p = 0.000), and 1.359 (p = 0.006), respectively. In this study population, obesity and elevated UA work together to increase the risk of renal injury. In vitro experiments indicate that reactive oxygen species (ROS) production increased with UA treatment in human renal glomerular endothelial cells (HRGECs), while endothelial nitric oxide synthase (eNOS) production level dropped. UA also increased monocyte chemotactic protein-1 (MCP-1) expression and nuclear factor kappa B (NF-κB) activation. Taken together, our results indicate that high concentrations of UA lead to endothelial dysfunction through the activation of the inflammatory response and induction of oxidative stress, even in non-obese T2DM patients.
Collapse
Affiliation(s)
- Yongmei Li
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xing Fan
- Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Chunjun Li
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xinyue Zhi
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Liyuan Peng
- Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Hongling Han
- Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Bei Sun
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| |
Collapse
|
49
|
|
50
|
Tassone EJ, Cimellaro A, Perticone M, Hribal ML, Sciacqua A, Andreozzi F, Sesti G, Perticone F. Uric Acid Impairs Insulin Signaling by Promoting Enpp1 Binding to Insulin Receptor in Human Umbilical Vein Endothelial Cells. Front Endocrinol (Lausanne) 2018; 9:98. [PMID: 29619007 PMCID: PMC5879120 DOI: 10.3389/fendo.2018.00098] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/28/2018] [Indexed: 12/22/2022] Open
Abstract
High levels of uric acid (UA) are associated with type-2 diabetes and cardiovascular disease. Recent pieces of evidence attributed to UA a causative role in the appearance of diabetes and vascular damage. However, the molecular mechanisms by which UA induces these alterations have not been completely elucidated so far. Among the mechanisms underlying insulin resistance, it was reported the role of a transmembrane glycoprotein, named either ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) or plasma cell antigen 1, which is able to inhibit the function of insulin receptor (IR) and it is overexpressed in insulin-resistant subjects. In keeping with this, we stimulated human umbilical vein endothelial cells (HUVECs) with insulin and UA to investigate the effects of UA on insulin signaling pathway, testing the hypothesis that UA can interfere with insulin signaling by the activation of ENPP1. Cultures of HUVECs were stimulated with insulin, UA and the urate transporter SLC22A12 (URAT1) inhibitor probenecid. Akt and endothelial nitric oxide synthase (eNOS) phosphorylation levels were investigated by immunoblotting. ENPP1 binding to IR and its tyrosine phosphorylation levels were tested by immunoprecipitation and immunoblotting. UA inhibited insulin-induced Akt/eNOS axis. Moreover, UA induced ENPP1 binding to IR that resulted in an impairment of insulin signaling cascade. Probenecid reverted UA effects, suggesting that UA intracellular uptake is required for its action. In endothelial cells, UA directly interferes with insulin signaling pathway at receptor level, through ENPP1 recruitment. This evidence suggests a new molecular model of UA-induced insulin resistance and vascular damage.
Collapse
Affiliation(s)
- Eliezer J. Tassone
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Antonio Cimellaro
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Maria Perticone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
- *Correspondence: Maria Perticone,
| | - Marta L. Hribal
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, Magna Græcia University, Catanzaro, Italy
| |
Collapse
|