1
|
Ouyang J, Wang H, Gan Y, Huang J. Uric acid mediates kidney tubular inflammation through the LDHA/ROS/NLRP3 pathway. Clin Exp Hypertens 2024; 46:2424834. [PMID: 39488824 DOI: 10.1080/10641963.2024.2424834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
PURPOSE Hyperuricemia (HUA) is an important factor leading to chronic kidney disease (CKD). The kidney tubular inflammatory response is activated in HUA. This study aimed to investigate whether lactate dehydrogenase A (LDHA) is involved in mediating uric acid-induced kidney tubular inflammatory response. METHODS In vivo, an HUA mouse model was established by continuous intraperitoneal injection of potassium oxonate (PO) for one week. A total of 18 C57BL/6J male adult mice were divided into three groups: control group, HUA group, and HUA+oxamate group, with six mice in each group. Oxamate was intraperitoneally injected into the mice one hour after PO injection. In vitro, an HUA model was simulated by stimulating HK-2 cells with uric acid. Oxamate and tempol inhibited LDHA and reactive oxygen species (ROS) in HK-2 cells. RESULTS In HUA mice, blood uric acid levels were significantly elevated. LDHA in kidney tubular cells was significantly increased in both in vivo and in vitro HUA models, accompanied by an increase in kidney tubular inflammation and ROS. Mechanistically, LDHA mediates uric acid-induced inflammation to kidney tubular cells through the ROS/NLRP3 pathway. Pharmacologic inhibition of LDHA or ROS in kidney tubular cells can significantly ameliorate inflammation response caused by uric acid. CONCLUSIONS LDHA in kidney tubular cells significantly was increased in HUA models. LDHA mediates kidney inflammation response induced by uric acid through the ROS/NLRP3 pathway. This study may provide a new intervention target for preventing kidney tubular inflammation caused by uric acid.
Collapse
Affiliation(s)
- Jun Ouyang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hui Wang
- School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yumei Gan
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiangnan Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
Zhu B, Li F, Zhang W, Zhao S, Song N, Jin S, Shen Z, Lu Y, Li Y, Liu H. Disparity of serum uric acid threshold for CKD among hypertensive and non-hypertensive individuals. Ren Fail 2024; 46:2301041. [PMID: 38425055 PMCID: PMC10911134 DOI: 10.1080/0886022x.2023.2301041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/27/2023] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION Hypertension and rising serum uric acid (sUA) played a pivotal role in the development of Chronic Kidney Disease (CKD). This study investigates the interactive effect of sUA and hypertension on CKD and identifies the optimal threshold of sUA among individuals with and without hypertension in the Chinese community population. MATERIALS AND METHODS The study included 4180 individuals aged 45-85 years, derived from the China Health and Retirement Longitudinal Study (CHARLS) between 2011 and 2015. Additionally, a hospital-based study enrolled subjects in the Department of Nephrology at Zhongshan Hospital, China from January 1, 2019, to December 31, 2021. The interaction effect analysis were used to assess the impact of sUA and hypertension on CKD. We also compared the distribution of sUA and the CKD risk in community populations, distinguishing between those with and without hypertension. For the hospital-based population, kidney injury was marked by a KIM-1 positive area. RESULTS Our results indicate a higher prevalence of CKD in the community population with hypertension (10.2% vs. 3.9%, p < .001). A significant additive synergistic effects of the sUA and hypertension on the CKD risk were found. When the sUA level was < 4.55 mg/dL in the hypertensive population and < 5.58 mg/dL in the non-hypertensive population, the risk of CKD was comparable (p = .809). In the propensity score matched (PSM) population, the result remained roughly constant. CONCLUSION Therefore, even moderate levels of sUA was associated with a higher risk of CKD in middle-aged hypertensive patients, who warrant stricter sUA control.
Collapse
Affiliation(s)
- Bowen Zhu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Fang Li
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Weidong Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Shi Jin
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Ziyan Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Yufei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Yang Li
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Hong Liu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| |
Collapse
|
3
|
Jung I, Lee DY, Chung SM, Park SY, Yu JH, Moon JS, Seo JA, Han K, Kim NH. Impact of Chronic Kidney Disease and Gout on End-Stage Renal Disease in Type 2 Diabetes: Population-Based Cohort Study. Endocrinol Metab (Seoul) 2024; 39:748-757. [PMID: 39212035 PMCID: PMC11525699 DOI: 10.3803/enm.2024.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGRUOUND We examined the impact of gout on the end-stage renal disease (ESRD) risk in patients with type 2 diabetes mellitus (T2DM) and determined whether this association differs according to chronic kidney disease (CKD) status. METHODS Using the Korean National Health Insurance Service, this nationwide cohort study enrolled 847,884 patients with T2DM who underwent health checkups in 2009. Based on the presence of CKD (estimated glomerular filtration rate <60 mL/min/1.73 m2) and gout (two outpatient visits or one hospitalization within 5 years), patients were classified into four groups: CKD-Gout-, CKD- Gout+, CKD+Gout-, and CKD+Gout+. Patients with incident ESRD were followed up until December 2018. RESULTS Among 847,884 patients, 11,825 (1.4%) experienced progression to ESRD. ESRD incidence increased in the following order: 0.77 per 1,000 person-years (PY) in the CKD-Gout- group, 1.34/1,000 PY in the CKD-Gout+ group, 8.20/1,000 PY in the CKD+Gout- group, and 23.06/1,000 PY in the CKD+Gout+ group. The presence of gout modified the ESRD risk in a status-dependent manner. Hazard ratios (HR) were 1.49 (95% confidence interval [CI], 1.32 to 1.69) and 2.24 (95% CI, 2.09 to 2.40) in patients without and with CKD, respectively, indicating a significant interaction (P<0.0001). The CKD+Gout+ group had a markedly higher risk of developing ESRD (HR, 18.9; 95% CI, 17.58 to 20.32) than the reference group (CKD-Gout-). CONCLUSION Gout substantially enhances the risk of ESRD, even in the absence of CKD. Concurrent CKD and gout synergistically increase the risk of ESRD. Therefore, physicians should carefully screen for hyperuricemia to prevent progression to ESRD.
Collapse
Affiliation(s)
- Inha Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Seung Min Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - So Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Ji Hee Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Jun Sung Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Ji A Seo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Nan Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| |
Collapse
|
4
|
Liu F, Bai Y, Wan Y, Luo S, Zhang L, Wu X, Chen R, Yin Z, Xie Y, Guo P. DaiTongXiao improves gout nephropathy by inhibiting inflammatory response through the TLR4/MyD88/NF-κB pathway. Front Pharmacol 2024; 15:1447241. [PMID: 39170709 PMCID: PMC11336418 DOI: 10.3389/fphar.2024.1447241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction: Gouty nephropathy (GN) arises from factors like excessive purine intake, metabolic disorders or abnormal synthesis, and uric acid hypersaturation in the blood, leading to urate crystal deposition in kidney tissue. DaiTongXiao (DTX) is a remedy used by the Dai people of China. It shows efficacy in lowering uric acid levels and exhibits anti-inflammatory and kidney-protective properties. Methods: A GN rat model was induced using adenine and potassium oxonate. Following DTX administration, various parameters were assessed in urine, serum, and kidney tissue. Western blot analysis evaluated TLR4/MyD88/NF-κB signaling proteins, while immunofluorescence examined NF-κB nuclear expression. Results: DTX treatment improved kidney morphology, increased body weight, and kidney index and enhanced urinary levels of blood urea nitrogen (Bun), 24-h urinary protein, uric acid (UA), and allantoin in GN rats, reducing UA, Bun, creatinine (Cre), cystatin C (CysC), serum amyloid A (SAA), α1-microglobulin (MG), and β2-MG in serum analysis. Renal tissue assessments showed decreased xanthine oxidase (XOD), hydroxyproline (Hyp), α-smooth muscle actin (α-SMA), and collage type Ⅳ (COL-Ⅳ). Kidney damage severity was notably reduced. DTX lowered serum inflammatory factors like interleukin (IL) -18, tumor necrosis factor-α (TNF-α), C-reactive protein (CRP), transforming growth factor-β1 (TGF-β1), and IL-1β in the rat serum, reducing chemokine monocyte chemoattractant protein-1 (MCP-1) and adhesion factor vascular cell adhesion molecule-1(VCAM-1). Western blotting demonstrated the downregulation of TLR4/MyD88/NF-κB pathway proteins, and immunofluorescence revealed reduced NF-κB expression in renal tissue. Discussion: DTX exhibits significant anti-GN effects by modulating TLR4/MyD88/ NF-κB pathway protein expression, reducing inflammatory factor release, and inhibiting GN progression.
Collapse
Affiliation(s)
- Feifan Liu
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yuanmei Bai
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yan Wan
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Shifang Luo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Linao Zhang
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Xue Wu
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Rong Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zili Yin
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yuhuan Xie
- College of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Peixin Guo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
5
|
Kang N, Ji Z, Li Y, Gao J, Wu X, Zhang X, Duan Q, Zhu C, Xu Y, Wen L, Shi X, Liu W. Metabolite-derived damage-associated molecular patterns in immunological diseases. FEBS J 2024; 291:2051-2067. [PMID: 37432883 DOI: 10.1111/febs.16902] [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: 11/25/2022] [Revised: 06/05/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
Damage-associated molecular patterns (DAMPs) are typically derived from the endogenous elements of necrosis cells and can trigger inflammatory responses by activating DAMPs-sensing receptors on immune cells. Failure to clear DAMPs may lead to persistent inflammation, thereby contributing to the pathogenesis of immunological diseases. This review focuses on a newly recognized class of DAMPs derived from lipid, glucose, nucleotide, and amino acid metabolic pathways, which are then termed as metabolite-derived DAMPs. This review summarizes the reported molecular mechanisms of these metabolite-derived DAMPs in exacerbating inflammation responses, which may attribute to the pathology of certain types of immunological diseases. Additionally, this review also highlights both direct and indirect clinical interventions that have been explored to mitigate the pathological effects of these DAMPs. By summarizing our current understanding of metabolite-derived DAMPs, this review aims to inspire future thoughts and endeavors on targeted medicinal interventions and the development of therapies for immunological diseases.
Collapse
Affiliation(s)
- Na Kang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhenglin Ji
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Yuxin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Ji Gao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Xinfeng Wu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, and College of Clinical Medical of Henan University of Science and Technology, Luoyang, China
| | - Xiaoyang Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Qinghui Duan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Can Zhu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Yue Xu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Luyao Wen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, and College of Clinical Medical of Henan University of Science and Technology, Luoyang, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, the First Affiliated Hospital, and College of Clinical Medical of Henan University of Science and Technology, Luoyang, China
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
6
|
Yuan HJ, Jiao HC, Liu XJ, Hao H, Liu Y, Xue YT, Li Y. Association of Serum Uric Acid with Non-Valvular Atrial Fibrillation: A Retrospective Study in China. Int J Gen Med 2024; 17:1533-1543. [PMID: 38680194 PMCID: PMC11048212 DOI: 10.2147/ijgm.s458089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024] Open
Abstract
Purpose The association between serum uric acid (SUA) and atrial fibrillation (AF) has been widely focused on and studied in recent years. However, the exact association between SUA and AF is unclear, and the effect of gender on the association between SUA levels and AF has been controversial. This study aimed to investigate the association between SUA levels and non-valvular AF (NVAF) and the potential effect of gender on it. Patients and Methods A total of 866 NVAF patients (463 males, age 69.44 ± 8.07 years) and 646 sex-matched control patients in sinus rhythm, with no history of arrhythmia were included in this study. t-test, ANOVA, and chi-square test were used for baseline data analysis. The receiver operating characteristic curve, logistic regression and Pearson correlation analysis were used for correlation analysis. Results Compared to controls, NVAF patients exhibited higher SUA (P<0.001). After adjusting for confounders of NVAF, SUA remained significantly associated with NVAF, regardless of gender (OR= 1.31, 95% CI 1.18-1.43, P<0.001). SUA demonstrated higher predictability and sensitivity in predicting the occurrence of female NVAF compared to male (area under the curve was 0.68 (95% CI 0.64-0.72, P<0.001), sensitivity 87.3%), with the optimal cut-off point identified as 5.72 mg/dL. Furthermore, SUA levels correlated with APOA1, Scr and NT-proBNP in NVAF patients. SUA levels varied significantly among NVAF subtypes. Conclusion High SUA levels were independently associated with NVAF, regardless of gender. SUA exhibited higher predictability and sensitivity in predicting the occurrence of NVAF in females compared to males. High SUA levels may affect other NVAF-related factors and participate in the pathophysiological process of NVAF.
Collapse
Affiliation(s)
- Hua-Jing Yuan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Hua-Chen Jiao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Xiu-Juan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Hao Hao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Yang Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Yi-Tao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, 250014, People’s Republic of China
| |
Collapse
|
7
|
Liu H, Chen Z, Liu M, Li E, Shen J, Wang J, Liu W, Jin X. The Terminalia chebula Retz extract treats hyperuricemic nephropathy by inhibiting TLR4/MyD88/NF-κB axis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117678. [PMID: 38159820 DOI: 10.1016/j.jep.2023.117678] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hyperuricemic nephropathy (HN) is a renal injury caused by hyperuricemia and is the main cause of chronic kidney disease and end-stage renal disease. ShiWeiHeZiSan, which is composed mainly of components of Terminalia chebula Retz. And is recorded in the Four Medical Tantras, is a typical traditional Tibetan medicinal formula for renal diseases. Although T. chebula has been reported to improve renal dysfunction and reduce renal cell apoptosis, the specific mechanism of the nephroprotective effects of T. chebula on HN is still unclear. AIM OF THE STUDY This study was conducted to evaluate the effects and specific mechanism of T. chebula extract on HN through network pharmacology and in vivo and in vitro experiments. MATERIALS AND METHODS Potassium oxalate (1.5 g/kg) and adenine (50 mg/kg) were combined for oral administration to establish the HN rat model, and the effects of T. chebula extract on rats in the HN model were evaluated by renal function indices and histopathological examinations. UPLC-Q-Exactive Orbitrap/MS analysis was also conducted to investigate the chemical components of T. chebula extract, and the potential therapeutic targets of T. chebula in HN were predicted by network pharmacology analysis. Moreover, the activation of potential pathways and the expression of related mRNAs and proteins were further observed in HN model rats and uric acid-treated HK-2 cells. RESULTS T. chebula treatment significantly decreased the serum uric acid (SUA), blood urea nitrogen (BUN) and serum creatinine (SCr) levels in HN rats and ameliorated renal pathological injury and fibrosis. A total of 25 chemical components in T. chebula extract were identified by UPLC-Q-Exactive Orbitrap/MS analysis, and network pharmacology analysis indicated that the NF-κB pathway was the potential pathway associated with the therapeutic effects of T. chebula extract on HN. RT‒PCR analysis, immunofluorescence staining and ELISA demonstrated that the mRNA and protein levels of TLR4 and MyD88 were significantly decreased in the renal tissue of HN rats after treatment with T. chebula extract at different concentrations, while the phosphorylation of P65 and the secretion of TNF-α and IL-6 were significantly inhibited. The results of in vitro experiments showed that T. chebula extract significantly decreased the protein levels of TLR4, MyD88, p-IκBα and p-P65 in uric acid-treated HK-2 cells and inhibited the nuclear translocation of p65 in these cells. In addition, the expression of inflammatory factors (IL-1β, IL-6 and TNF-α) and fibrotic genes (α-SMA and fibronectin) was significantly downregulated by T. chebula extract treatment, while E-cadherin expression was significantly upregulated. CONCLUSION T. chebula extract exerts nephroprotective effects on HN, such as anti-inflammatory effects and fibrosis improvement, by regulating the TLR4/MyD88/NF-κB axis, which supports the general use of T. chebula in the management of HN and other chronic kidney diseases.
Collapse
Affiliation(s)
- Hao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China; School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China
| | - Zhiyu Chen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China; School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China
| | - Meng Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China; School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China
| | - Ertong Li
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China; School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China
| | - Juan Shen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China; School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China
| | - Jie Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China; School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China
| | - Wenbin Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China; School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China.
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China; School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou Guangdong 510006, PR China.
| |
Collapse
|
8
|
Fan Z, Li Z, Guo A, Li Y. The association of low serum uric acid with mortality in older people is modified by kidney function: National Health and Nutrition Examination Survey (NHANES) 1999-2018. BMC Nephrol 2024; 25:108. [PMID: 38504168 PMCID: PMC10953214 DOI: 10.1186/s12882-024-03546-6] [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: 05/17/2023] [Accepted: 03/14/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND In older individuals, the role of low serum uric acid (SUA) as risk factor for mortality is debated. We therefore studied whether SUA levels, particularly low SUA concentrations, are associated with all-cause and cardiovascular (CV) mortality in older population, and to clarify potential effect modification of kidney function. METHODS We identified 14,005 older people in National Health and Nutrition Examination Survey (NHANES) data from 1999 to 2018. SUA was measured only at baseline. The relationship between SUA and mortality was assessed using Cox proportional hazards models and restricted cubic spline Cox regression stratified by the estimated glomerular filtration rate (eGFR). RESULTS During mean 8.3 years of follow-up, 4852 all-cause death and 1602 CV death were recorded. A significant U-shaped association was observed between SUA with all-cause mortality, with the lowest risk concentration of 5.5 mg/dL. Comparing to the reference group (5 to 7 mg/dL), the HR of 2 to < 5 mg/dL group was 1.11 (1.03-1.21) and 1.14 (1.00-1.30). This relationship was more pronounced in participants with an eGFR ≥ 60 ml/min/1.73m2 (HR, 1.16; 95%CI, 1.06-1.28). This situation similarly occurred in Urine protein negative group (HR, 1.14; 95%CI, 1.04-1.25). CONCLUSIONS Low SUA concentrations are associated with an increased risk in all-cause and CV mortality among older participants. Extremely low SUA concentrations are especially undesirable, especially in the older adults with normal kidney function.
Collapse
Affiliation(s)
- Zhongcheng Fan
- Department of Osteology, Haikou Municipal People's Hospital and Central South University Xiangya Medical College Affiliated Hospital, Haikou, China
| | - Zhongju Li
- Division of Ultrasonography, Haikou Municipal People's Hospital and Central South University Xiangya Medical College Affiliated Hospital, Haikou, China
| | | | - Yang Li
- Division of Nephrology and Rheumatology, Haikou Municipal People's Hospital and Central South University Xiangya Medical College Affiliated Hospital, 43 Renmin Ave, Haikou, 570208, China.
| |
Collapse
|
9
|
Asahina Y, Sakaguchi Y, Oka T, Hattori K, Kawaoka T, Doi Y, Yamamoto R, Matsui I, Mizui M, Kaimori JY, Isaka Y. Association between urinary uric acid excretion and kidney outcome in patients with CKD. Sci Rep 2024; 14:5119. [PMID: 38429496 PMCID: PMC10907602 DOI: 10.1038/s41598-024-55809-9] [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: 09/11/2023] [Accepted: 02/28/2024] [Indexed: 03/03/2024] Open
Abstract
Inhibiting tubular urate reabsorption may protect the kidney from urate-induced tubular injury. However, this approach may promote intratubular uric acid crystallization, especially in acidified urine, which could be toxic to the kidney. To assess how tubular urate handling affects kidney outcomes, we conducted a retrospective cohort study including 1042 patients with estimated glomerular filtration rates (eGFR) of 15-60 mL/min/1.73 m2. The exposures were fractional excretion of uric acid (FEUA) and urinary uric acid-to-creatinine ratio (UUCR). The kidney outcome was defined as a halving of eGFR from baseline or initiating kidney replacement therapy. The median FEUA and UUCR were 7.2% and 0.33 g/gCre, respectively. During a median follow-up of 1.9 years, 314 kidney outcomes occurred. In a multivariate Cox model, the lowest FEUA quartile exhibited a 1.68-fold higher rate of kidney outcome than the highest FEUA quartile (95% confidence interval, 1.13-2.50; P = 0.01). Similarly, lower UUCR was associated with a higher rate of kidney outcome. Notably, patients in the highest quartile of FEUA and UUCR were at the lowest risk of kidney outcome even among those with aciduria. In conclusion, lower FEUA and UUCR were associated with a higher risk of kidney failure, suggesting that increased urate reabsorption is harmful to the kidney.
Collapse
Affiliation(s)
- Yuta Asahina
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yusuke Sakaguchi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Tatsufumi Oka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koki Hattori
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takayuki Kawaoka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yohei Doi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryohei Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
- Health and Counseling Center, Osaka University, Toyonaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jun-Ya Kaimori
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
10
|
Lu H, Zhang H, Wu Z, Li L. Microbiota-gut-liver-brain axis and hepatic encephalopathy. MICROBIOME RESEARCH REPORTS 2024; 3:17. [PMID: 38841407 PMCID: PMC11149093 DOI: 10.20517/mrr.2023.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 06/07/2024]
Abstract
Hepatic encephalopathy (HE) is a clinical manifestation of neurological and psychiatric abnormalities that are caused by complications of liver dysfunction including hyperammonemia, hyperuricemia, and portal hypertension. Accumulating evidence suggests that HE could be reversed through therapeutic modifications of gut microbiota. Multiple preclinical and clinical studies have indicated that gut microbiome affects the physiological function of the liver, such as the regulation of metabolism, secretion, and immunity, through the gut-liver crosstalk. In addition, gut microbiota also influences the brain through the gut-brain crosstalk, altering its physiological functions including the regulation of the immune, neuroendocrine, and vagal pathways. Thus, key molecules that are involved in the microbiota-gut-liver-brain axis might be able to serve as clinical biomarkers for early diagnosis of HE, and could be effective therapeutic targets for clinical interventions. In this review, we summarize the pathophysiology of HE and further propose approaches modulating the microbiota-gut-liver-brain axis in order to provide a comprehensive understanding of the prevention and potential clinical treatment for HE with a microbiota-targeted therapy.
Collapse
Affiliation(s)
| | | | | | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| |
Collapse
|
11
|
Xu G, Wu L, Yang H, Liu T, Tong Y, Wan J, Han B, Zhou L, Hu X. Eupatilin inhibits xanthine oxidase in vitro and attenuates hyperuricemia and renal injury in vivo. Food Chem Toxicol 2024; 183:114307. [PMID: 38052408 DOI: 10.1016/j.fct.2023.114307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
Uric acid (UA) is the final metabolite of purines in the liver that can cause hyperuricemia at high levels. The kidneys are the main excretory organs for UA. The excessive accumulation of UA in the kidneys causes the development of hyperuricemia that often leads to renal injury. Eupatilin (Eup) is a flavonoid natural product that possesses various pharmacological properties such as antioxidant, anti-cancer, and anti-inflammatory. We were interested in exploring the potential role of Eup in lowering UA and nephroprotective. We initially investigated the effects of Eup on xanthin oxidase (XOD) activity in vitro, followed by investigating its ability to lower UA levels, anti-inflammatory effects, nephroprotective effects, and the underlying mechanisms using hyperuricemia rats sustained at high UA level. The results showed that Eup had an inhibitory effect on XOD activity in vitro and significantly reduced serum UA, creatinine, BUN, IL-1β and IL-6 levels in hyperuricemic rats, ameliorating inflammation, renal oxidative stress and pathological injury. Furthermore, Eup inhibited ADA and XOD enzyme activities in the liver and serum and modulated GLUT9, URAT1 and ABCG2 protein expression in the kidneys and ileum. Our findings provide a scientific basis for suggesting Eup as an option for a potential treatment for hyperuricemia.
Collapse
Affiliation(s)
- Guitao Xu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Lele Wu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Hongxuan Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Tianfeng Liu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Ying Tong
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jiliang Wan
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Bin Han
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Lin Zhou
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xuguang Hu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
12
|
Zhou J, Wang C, Zhang X, Wu Z, Wu Y, Li D, Gao J. Shizhifang ameliorates pyroptosis of renal tubular epithelial cells in hyperuricemia through inhibiting NLRP3 inflammasome. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116777. [PMID: 37311502 DOI: 10.1016/j.jep.2023.116777] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese Medicine (TCM) Compound Shizhifang (SZF), consisting of the seeds of four Chinese herbs, has been used in Shanghai Shuguang Hospital in China for more than 20 years and has proven its clinical safety and efficacy in lowering uric acid and protecting kidney function. AIM OF THE STUDY Hyperuricemia (HUA)-induced pyroptosis of renal tubular epithelial cells serves as a significant cause of tubular damage. SZF proves to be effective in alleviating renal tubular injury and inflammation infiltration of HUA. However, the inhibiting effect of SZF on pyroptosis in HUA still remains elusive. This study aims to verify whether SZF could ameliorate pyroptosis in tubular cells induced by uric acid (UA). MATERIALS AND METHODS Quality control analysis and chemical and metabolic identification for SZF and SZF drug serum were performed by using UPLC-Q-TOF-MS. In vitro, human renal tubular epithelial cells (HK-2) stimulated by UA were treated with SZF or NLRP3 inhibitor (MCC950). HUA mouse models were induced by intraperitoneal injection of potassium oxonate (PO). Mice were treated with SZF, allopurinol or MCC950. We focused on evaluated the effect of SZF on the NLRP3/Caspase-1/GSDMD pathway, renal function, pathologic structure and inflammation. RESULTS SZF significantly restrained the activation of the NLRP3/Caspase-1/GSDMD pathway in vitro and in vivo induced by UA. SZF was better than allopurinol and MCC950 in reducing pro-inflammatory cytokine levels, attenuating tubular inflammatory injury, inhibiting interstitial fibrosis and tubular dilation, maintaining tubular epithelial cell function, and protecting kidney. Furthermore, 49 chemical compounds of SZF and 30 metabolites in serum after oral administration were identified. CONCLUSIONS SZF inhibits UA-induced renal tubular epithelial cell pyroptosis via by targeting NLRP3 to inhibit tubular inflammatory and prevent the progression of HUA-induced renal injury effectively.
Collapse
Affiliation(s)
- Jiabao Zhou
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Road ZhangHeng, Shanghai, China
| | - Chuanxu Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Road ZhangHeng, Shanghai, China
| | - Xuming Zhang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Road ZhangHeng, Shanghai, China
| | - Zhiyuan Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Road ZhangHeng, Shanghai, China
| | - Yansheng Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Road ZhangHeng, Shanghai, China
| | - Dongdong Li
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Road ZhangHeng, Shanghai, China
| | - Jiandong Gao
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Road ZhangHeng, Shanghai, China.
| |
Collapse
|
13
|
Wang X, Li X, Wang H, Chen M, Wen C, Huang L, Zhou M. All-cause and specific mortality in patients with gout: A systematic review and meta-analysis. Semin Arthritis Rheum 2023; 63:152273. [PMID: 37832433 DOI: 10.1016/j.semarthrit.2023.152273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/03/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
OBJECTIVES This meta-analysis aims to examine the general mortality risk and specific mortality risk of gout, as the incidence of the condition is on the rise but information on mortality rates remains uncertain. METHOD The researchers conducted a search of published cohort studies on gout and mortality using Medical subject headings and keywords in PubMed, EMBASE, and Cochrane Library databases from inception to September 2022. The quality of study was evaluated using the NOS scale. Statistical analysis was performed using STATA software (version 16.0). Publication bias was assessed using funnel plots and Egger's test. RESULT This meta-analysis included 11 cohort studies (2010-2022), covering 14,854,490 people with a follow-up time of 1.66-16 years. The pooled analysis shows increased risk of overall mortality [HR=1.23, 95 % CI (1.13-1.35), I2=96.4 %, P<0.001], cardiovascular mortality [HR=1.29, 95 % CI (1.13-1.48), I2=98.5 %, P<0.001], infection mortality [HR=1.24, 95 % CI (1.04-1.47), I2=88.5 %, P = 0.019], and digestive system disease mortality [HR=1.42, 95 % CI (1.13-1.80), I2=91.7 %, P = 0.003] in gout. Sensitivity and subgroup analysis support the findings, and publication bias was not evident. CONCLUSION The findings from our meta-analysis indicate that gout is associated with an increased risk of all-cause mortality, as well as mortality related to cardiovascular disease, infections, and digestive system diseases. This has important implications for clinical practice, nursing care of patients with gout, and guidance on lifestyle modifications to prevent adverse outcomes such as cardiovascular events, infections, and digestive disorders.
Collapse
Affiliation(s)
- Xueyan Wang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, China
| | - Xuanlin Li
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, China
| | - Hongli Wang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, China
| | - Min Chen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, China
| | - Chengping Wen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, China
| | - Lin Huang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, China.
| | - Mingqian Zhou
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Science, Zhejiang Chinese Medical University, Binwen Road, Binjiang District, Hangzhou, China.
| |
Collapse
|
14
|
Wang Q, Lu X, Xu L, Liang H. Gender variations in the impact of hyperuricemia on thyroid disorders. Endocr Res 2023; 48:77-84. [PMID: 37382510 DOI: 10.1080/07435800.2023.2231092] [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: 01/07/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023]
Abstract
This study aimed to examine the impact of hyperuricemia on various thyroid disorders with emphasized focus on differences resulting from different genders. 16094 adults aged ≥18 years were enrolled in this cross-sectional study using a randomized stratified sampling strategy. Clinical data including thyroid function and antibodies, uric acid, and anthropometric measurements were measured. Multivariable logistic regression was used to determine the association between hyperuricemia and thyroid disorders. Women who have hyperuricemia are at a significantly increased risk of developing hyperthyroidism. Women's risk of overt hyperthyroidism and Graves' disease may be markedly increased by hyperuricemia. Men with hyperuricemia did not differ significantly in their chance of acquiring any thyroid disorders.
Collapse
Affiliation(s)
- Qiang Wang
- Medical Education Administrate Office, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Xixuan Lu
- Department of Endocrinology, Cardiovascular and Cerebrovascular Disease Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Li Xu
- Department of Radiology, The 942th Hospital of the People's Liberation Army Joint Logistics Support Force, Yinchuan, China
| | - Haiyan Liang
- Department of Endocrinology, Cardiovascular and Cerebrovascular Disease Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
15
|
Dai R, Peng C, Sang T, Cheng M, Wang Y, Zhang L. Construction and validation of a predictive model for the risk of peritoneal dialysis-associated peritonitis after peritoneal dialysis catheterization. Front Med (Lausanne) 2023; 10:1193754. [PMID: 37790129 PMCID: PMC10543278 DOI: 10.3389/fmed.2023.1193754] [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: 03/25/2023] [Accepted: 09/01/2023] [Indexed: 10/05/2023] Open
Abstract
Aim To construct and validate a risk prediction model for the development of peritoneal dialysis-associated peritonitis (PDAP) in patients undergoing peritoneal dialysis (PD). Methods This retrospective analysis included patients undergoing PD at the Department of Nephrology, the First Affiliated Hospital of Anhui University of Chinese Medicine, between January 2016 and January 2021. Baseline data were collected. The primary study endpoint was PDAP occurrence. Patients were divided into a training cohort (n = 264) and a validation cohort (n = 112) for model building and validation. Least Absolute Shrinkage and Selection Operator (LASSO) regression was applied to optimize the screening variables. Predictive models were developed using multifactorial logistic regression analysis with column line plots. Receiver operating characteristic (ROC) curves, calibration curves, and Hosmer-Lemeshow goodness-of-fit tests were used to verify and evaluate the discrimination and calibration of the prediction models. Decision curve analysis (DCA) was used to assess the clinical validity of the prediction models. Results Five potential predictors of PDAP after PD catheterization were screened using LASSO regression analysis, including neutrophil-to-lymphocyte ratio (NLR), serum ALBumin (ALB), uric acid (UA), high sensitivity C-reactive protein (hsCRP), and diabetes mellitus (DM). Predictive models were developed by multi-factor logistic regression analysis and plotted in columns. The area under the ROC curve (AUC) values were 0.891 (95% confidence interval [CI]: 0.829-0.844) and 0.882 (95% CI: 0.722-0.957) for the training and validation cohorts, respectively. The Hosmer-Lemeshow test showed a good fit (p = 0.829 for the training cohort; p = 0.602 for the validation cohort). The DCA curves indicated that the threshold probabilities for the training and validation cohorts were 4-64% and 3-90%, respectively, predicting a good net gain for the clinical model. Conclusion NLR, ALB, UA, hsCRP, and DM are independent predictors of PDAP after PD catheterization. The column line graph model constructed based on the abovementioned factors has good discriminatory and calibrating ability and helps to predict the risk of PDAP after PD catheterization.
Collapse
Affiliation(s)
- Rong Dai
- Department of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chuyi Peng
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Tian Sang
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Meng Cheng
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Yiping Wang
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Lei Zhang
- Department of Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
16
|
Cimmino G, Gallinoro E, di Serafino L, De Rosa G, Sugraliyev A, Golino P, Cirillo P. Uric acid plasma levels are associated with C-reactive protein concentrations and the extent of coronary artery lesions in patients with acute coronary syndromes. Intern Emerg Med 2023; 18:1751-1757. [PMID: 37466806 DOI: 10.1007/s11739-023-03360-2] [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: 03/31/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023]
Abstract
Many studies have pointed out that inflammation plays a pivotal role in pathophysiology of acute coronary syndromes (ACS) because several inflammatory molecules impair the endothelial functions in the coronary circulation and promote atherothrombotic events. Recently, many clinical/experimental evidences indicate that elevated plasma levels of uric acid (UA) might be considered a risk factor for developing ACS. It has been reported that elevated UA doses impair physiologic functions of endothelial cells, shifting them toward a pro atherothrombotic phenotype. In the present manuscript, we investigated the relationship between UA plasma levels, inflammatory burden, and extension of coronary atherosclerotic disease in patients with ACS. Patients with a clinical presentation of ACS (ST-elevated and non-ST-elevated myocardial infarction) admitted to the Vanvitelli Catheterization Laboratory at Monaldi Hospital in 2019, before the COVID-19 pandemia, were retrospectively analyzed. Biochemical profile, type of ACS presentation, as well as extension of coronary atherosclerosis were assessed. A total of 132 ACS patients were included in the analysis, and grouped into 3 tertiles according to the UA values (UA < 4.72 mg/dl, UA between 4.72 and 6.15 mg/dl, and UA > 6.15 mg/dl). Patients with UA plasma levels ≥ 6.15 mg/dL showed higher levels of C-reactive protein (mean of 5.1 mg/dL) as compared to patients with lower UA plasma levels. Moreover, the former group of patients showed higher levels of cardiac troponin and CPK, and presented more often with multivessel disease and complex coronary stenosis (type C of Ellis classification). Even though monocentric and with limited sample size, the present study shows that plasma levels of UA and hs-CRP are elevated in ACS patients and are associated with a more severe coronary disease, suggesting a potential role of UA in the pathophysiology of acute coronary events.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Emanuele Gallinoro
- Cardiology Department, IRCCS Galeazzi-Sant'Ambrogio Hospital, University of Milan, Milan, Italy
| | - Luigi di Serafino
- Department of Advanced Biomedical Sciences, Section of Cardiology, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Gennaro De Rosa
- Department of Advanced Biomedical Sciences, Section of Cardiology, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Akhmetzhan Sugraliyev
- Department of Internal Disease, Kazakh National Medical University, Almaty, Kazakhstan
| | - Paolo Golino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, Section of Cardiology, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
17
|
Wojtasińska A, Frąk W, Lisińska W, Sapeda N, Młynarska E, Rysz J, Franczyk B. Novel Insights into the Molecular Mechanisms of Atherosclerosis. Int J Mol Sci 2023; 24:13434. [PMID: 37686238 PMCID: PMC10487483 DOI: 10.3390/ijms241713434] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Atherosclerosis is one of the most fatal diseases in the world. The associated thickening of the arterial wall and its background and consequences make it a very composite disease entity with many mechanisms that lead to its creation. It is an active process, and scientists from various branches are engaged in research, including molecular biologists, cardiologists, and immunologists. This review summarizes the available information on the pathophysiological implications of atherosclerosis, focusing on endothelium dysfunction, inflammatory factors, aging, and uric acid, vitamin D, and miRNA expression as recent evidence of interactions of the molecular and cellular elements. Analyzing new discoveries for the underlying causes of this condition assists the general research to improve understanding of the mechanism of pathophysiology and thus prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Armanda Wojtasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Weronika Frąk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Natalia Sapeda
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| |
Collapse
|
18
|
Abdulhafiz F, Mohammed A, Reduan MFH, Hamzah Z, Kari ZA, Téllez-Isaías G. Evaluation of anti-hyperuricemic effects of Alocasia longiloba Miq. (Keladi Candik) extracts in potassium oxonate induced rat model. Heliyon 2023; 9:e18069. [PMID: 37483701 PMCID: PMC10362144 DOI: 10.1016/j.heliyon.2023.e18069] [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: 05/17/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 07/25/2023] Open
Abstract
Hyperuricemia has become a significant public-health concern in recent years, and the available treatments have been reported to have an adverse side effect on patients. Alocasia longiloba has been used traditionally in Malaysia for treating gout, inflammation, and wounds. However, the plant has not been investigated for its effects on hyperuricemia. This study investigated the anti-hyperuricemic and anti-inflammatory effects of A. longiloba extracts in hyperuricemic rats induced by potassium oxonate (250 mg/kg body weight). Rats were given A. longiloba extracts or a standard drug for two-week, and blood and tissue samples were collected for analysis. Results show that A. longiloba extracts significantly reduced serum uric acid levels in hyperuricemic rats and inhibited xanthine oxidase (XOD) activity in the liver and kidney, which could be the mechanism underlying the urate-lowering effects. The extracts also significantly (p < 0.05) reduced the levels of proinflammatory cytokines (IL-18 and IL-1β) in serum samples and had hepatoprotective and nephroprotective effects in hyperuricemic rats. The study supports the use of A. longiloba as a complementary therapy for treating hyperuricemia.
Collapse
Affiliation(s)
- Ferid Abdulhafiz
- Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Kelantan, Malaysia
- Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa, 16100 Kota Bharu, Kelantan, Malaysia
- Department of Agricultural Sciences, Faculty of Applied Science, Lincoln University College, 47301 Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Arifullah Mohammed
- Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Kelantan, Malaysia
| | - Mohd Farhan Hanif Reduan
- Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa, 16100 Kota Bharu, Kelantan, Malaysia
| | - Zulhazman Hamzah
- Faculty of Earth Science, Universiti Malaysia Kelantan, Jeli 17600, Kelantan, Malaysia
| | - Zulhisyam Abdul Kari
- Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Kelantan, Malaysia
| | | |
Collapse
|
19
|
Wang X, Liu X, Gao Q, Gu X, Zhang G, Sheng Z, Wu T, Su Z, Wang W, Ye M. Gegen Qinlian Decoction treatment of asymptomatic hyperuricemia by targeting circadian immune function. Chin Med 2023; 18:77. [PMID: 37370132 DOI: 10.1186/s13020-023-00775-z] [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: 02/20/2023] [Accepted: 05/20/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The Gegen Qinlian Decoction (GGQLD) is a renowned traditional Chinese medicinal formula that has been used for centuries to effectively treat asymptomatic Hyperuricemia (HUA). This study aims to investigate the underlying mechanism of GGQLD's therapeutic effects on HUA. METHODS The study enrolled a total of 25 healthy participants and 32 middle-aged and elderly individuals with asymptomatic HUA. All asymptomatic HUA participants were treated with GGQLD. Venous blood samples were collected from all participants to isolate peripheral blood mononuclear cells (PBMCs), which were then analyzed for biological profiles using flow cytometry. Network pharmacology analysis was utilized to identify the potential pathways involved in the therapeutic effects of GGQLD. Transcriptomic patterns of cultured proximal tubule epithelial cells (PTECs) were evaluated via bulk RNA-seq, and critical differentially expressed genes (DEGs) were identified and verified through ELISA. Molecular docking and molecular dynamics (MD) simulation were employed to investigate the potential compounds in GGQLD that may be involved in treating HUA. RESULTS Network pharmacology analysis revealed that immune-related pathways might be involved in the therapeutic mechanism of GGQLD. RNA-seq analysis confirmed the involvement of innate lymphoid cell (ILC) development-related genes and clock genes. Polychromatic flow cytometric analysis demonstrated that GGQLD treatment reduced the proportion of ILC3s in total ILCs in asymptomatic HUA patients. ELISA results showed that GGQLD treatment reduced the levels of activating factors, such as ILC3-IL-18 and IL-1β, in the plasma of HUA patients. GGQLD was also found to regulate circadian clock gene expression in PBMCs to treat asymptomatic HUA. Furthermore, the interaction between 40 compounds in GGQLD and HDAC3 (Histone Deacetylase 3), NLRP3 (NOD-like receptor protein 3), RORA (RAR-related orphan receptor A), and REV-ERBα (nuclear receptor subfamily 1) revealed that GGQLD may regulate ILCs and clock genes to treat asymptomatic HUA. CONCLUSIONS The regulation of circadian clock gene expression and the proportion of ILC cells may be involved in the therapeutic effects of GGQLD on asymptomatic HUA patients.
Collapse
Affiliation(s)
- Xiaojun Wang
- Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, No 221West Yan-An Road, Shanghai, 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No 221 West Yan-An Road, Shanghai, 200040, China
| | - Xuanqi Liu
- Department of Respiratory and Critical Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Qiushuang Gao
- China Pharmaceutical University, Nanjing, 210009, China
| | - Xuchao Gu
- Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, No 221West Yan-An Road, Shanghai, 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No 221 West Yan-An Road, Shanghai, 200040, China
| | - Guannan Zhang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Zhiyuan Sheng
- Department of Urology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Tao Wu
- Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, No 221West Yan-An Road, Shanghai, 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No 221 West Yan-An Road, Shanghai, 200040, China
| | - Zheling Su
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Wenhao Wang
- Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, No 221West Yan-An Road, Shanghai, 200040, China.
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No 221 West Yan-An Road, Shanghai, 200040, China.
| | - Maoqing Ye
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, No 221 West Yan-An Road, Shanghai, 200040, China.
- Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China.
| |
Collapse
|
20
|
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
|
21
|
Amatya B, Yang S, Yu P, Vaz de Castro PA, Armando I, Zeng C, Felder RA, Asico LD, Jose PA, Lee H. Peroxiredoxin-4 and Dopamine D5 Receptor Interact to Reduce Oxidative Stress and Inflammation in the Kidney. Antioxid Redox Signal 2023; 38:1150-1166. [PMID: 36401517 PMCID: PMC10262345 DOI: 10.1089/ars.2022.0034] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 11/05/2022] [Accepted: 11/05/2022] [Indexed: 11/21/2022]
Abstract
Aims: Reactive oxygen species are highly reactive molecules generated in different subcellular compartments. Both the dopamine D5 receptor (D5R) and endoplasmic reticulum (ER)-resident peroxiredoxin-4 (PRDX4) play protective roles against oxidative stress. This study is aimed at investigating the interaction between PRDX4 and D5R in regulating oxidative stress in the kidney. Results: Fenoldopam (FEN), a D1R and D5R agonist, increased PRDX4 protein expression, mainly in non-lipid rafts, in D5R-HEK 293 cells. FEN increased the co-immunoprecipitation of D5R and PRDX4 and their colocalization, particularly in the ER. The efficiency of Förster resonance energy transfer was increased with FEN treatment measured with fluorescence lifetime imaging microscopy. Silencing of PRDX4 increased hydrogen peroxide production, impaired the inhibitory effect of FEN on hydrogen peroxide production, and increased the production of interleukin-1β, tumor necrosis factor (TNF), and caspase-12 in renal cells. Furthermore, in Drd5-/- mice, which are in a state of oxidative stress, renal cortical PRDX4 was decreased whereas interleukin-1β, TNF, and caspase-12 were increased, relative to their normotensive wild-type Drd5+/+ littermates. Innovation: Our findings demonstrate a novel relationship between D5R and PRDX4 and the consequent effects of this relationship in attenuating hydrogen peroxide production in the ER and the production of proinflammatory cytokines. This study provides the potential for the development of biomarkers and new therapeutics for renal inflammatory disorders, including hypertension. Conclusion: PRDX4 interacts with D5R to decrease oxidative stress and inflammation in renal cells that may have the potential for translational significance. Antioxid. Redox Signal. 38, 1150-1166.
Collapse
Affiliation(s)
- Bibhas Amatya
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | - Sufei Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Peiying Yu
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Molecular Physiology Research, Children's National Medical Center, Washington, District of Columbia, USA
| | - Pedro A.S. Vaz de Castro
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | - Ines Armando
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Molecular Physiology Research, Children's National Medical Center, Washington, District of Columbia, USA
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Robin A. Felder
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, Virginia, USA
| | - Laureano D. Asico
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Molecular Physiology Research, Children's National Medical Center, Washington, District of Columbia, USA
| | - Pedro A. Jose
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Molecular Physiology Research, Children's National Medical Center, Washington, District of Columbia, USA
- Department of Pharmacology/Physiology, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | - Hewang Lee
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, USA
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Molecular Physiology Research, Children's National Medical Center, Washington, District of Columbia, USA
| |
Collapse
|
22
|
Meng J, Tian J, Zhao Y, Li C, Yi Y, Zhang Y, Han J, Wang L, Pan C, Liu S, Liu C, Wang F, Tang X, Wang D, Qin S, Liang A. Ameliorative effect of cheqianzi decoction on hyperuricemia and kidney injury and underlying mechanism in rats. Heliyon 2023; 9:e15333. [PMID: 37123969 PMCID: PMC10130219 DOI: 10.1016/j.heliyon.2023.e15333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/19/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
Cheqianzi Decoction (CQD) is a Traditional Chinese Medicine (TCM) formula comprising four herbs and is recorded in the Ancient Materia Medica "Shengji Zonglu". Individually, these four herbs have been shown to reduce uric acid (UA) levels, to treat hyperuricemia (HUA), and alleviate kidney damage. However, the therapeutic efficacy of the CQD and related mechanism are not yet clear. In this study, high performance liquid chromatography (HPLC) analysis confirmed that the contents of the chemical components of the four herbal medicines were in accordance with the provisions of the Chinese Pharmacopoeia. A total of 99 potential targets were identified in the network pharmacology analysis of CQD, indicating its involvement in the regulation of inflammatory and apoptotic signaling pathways, and potential value for treating HUA and alleviating kidney injury. In vivo pharmacodynamic studies showed that compared with the Model group, significantly decreased levels of serum uric acid (SUA), serum creatinine (SCr), blood urea nitrogen (BUN) (all P < 0.05), and inflammatory factors (P < 0.01) were detected in the CQD group. Quantitative real-time PCR and Western blot analyses showed that compared with the Model group, adenosine triphosphate (ATP)-binding cassette efflux transporter G2 (ABCG2) expression in the CQD group was significantly upregulated (P < 0.01) at both the mRNA and protein levels, while mRNA expression of Caspase3 and NOD-like receptor family member 3 (NLRP3) (P < 0.05) and protein expression of NLRP3 (P < 0.01) were significantly downregulated. In conclusion, CQD promotes UA excretion by activating ABCG2, and induces inflammasome NLRP3-mediated reduction in inflammatory and apoptotic factors to achieve renal protection. Thus, our findings indicate the therapeutic potential of CQD in HUA with kidney injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Aihua Liang
- Corresponding author. Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimen Nei Ave, Beijing, 100700, China.
| |
Collapse
|
23
|
Chrysant SG. Association of hyperuricemia with cardiovascular diseases: current evidence. Hosp Pract (1995) 2023; 51:54-63. [PMID: 36730938 DOI: 10.1080/21548331.2023.2173413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The aim of the present study is to present a historical and unified perspective on the association of serum uric acid (SUA) in the cause of cardiovascular diseases (CVDs). The association of hyperuricemia (HUC) with CVD begun to be appreciated in the middle 1950s and early 1990s when clinical evidence was shown on the association of HUC with CVD. However, this association was disputed by several investigators including the Framingham group and by professional societies, like the American Heart Association and the American Society of Hypertension. This dispute was weakened or reversed by later studies, which showed a positive association of HUC with CVD, CHD, HF, CKD, and stroke, mediated by several risk factors, both molecular such as, oxidative stress, inflammatory stress, insulin resistance, and endothelial dysfunction, as well as clinical factors such as, atherosclerosis, hypertension, metabolic syndrome, and type 2 diabetes mellitus. The great majority of recent studies show a positive association of HUC with CVDs, and CKD. However, the cutoff of the damaging levels of SUA have not been established as yet. The European Society of Hypertension (ESH) Treatment Guidelines have proposed a cutoff level of SUA for CVD > 7 mg/dl for men and > 6 mg/dl for women. In contrast, the URRAH study has shown a SUA level of 4.7 mg/dl for all-cause mortality and 5.6 mg/dl for CV mortality. These levels are lower than the SUA levels proposed by the ESH, which are consistent with HUC. For a better understanding of this association, a Medline search of the English literature was conducted between 2015 and 2022 and 44 pertinent papers were selected. These papers together with collateral literature will be discussed in this review.
Collapse
Affiliation(s)
- Steven G Chrysant
- Department of Cardiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
24
|
Fang M, Li B, Li X, Wang Y, Zhuang Y. MicroRNA-29b regulates pyroptosis involving calcific aortic valve disease through the STAT3/SOCS1 pathway. Int J Cardiol 2023; 371:319-328. [PMID: 36064035 DOI: 10.1016/j.ijcard.2022.08.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 08/03/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND CAVD (calcific aortic valve disease) involves an inflammatory response similar to pyroptosis; therefore, we speculated that the progression of pyroptosis might be involved in the pathogenesis of CAVD. METHODS We first investigated the expression of pyroptosis related genes in human CAVD, non-CAVD control and AS (calcific aortic stenosis) tissues. We further confirmed these genes by using CAVD cell and mouse models. Finally, we explored the functional molecular mechanism in the cell model. RESULTS Our recent studies found that miR-29b plays an important role in CAVD, and we wanted to further address whether miR-29b is a key factor in the progression of pyroptosis related to CAVD. In this study, we found NLRP3 was highly expressed in CAVD patients and models. In contrast, SOCS1, a suppressor of NLRP3, showed reduced expression in CAVD. Furthermore, we found that ASC, Caspase-1, IL-1β, Cleaved IL-18 and p-JAK2 were all upregulated in the tissues of CAVD patients, suggesting the likelihood of activation of the inflammasome. Then, we found that miR-29b participated in the NLRP3-regulated CAVD pathway through its target gene STAT3 (signal transducer and activator of transcription 3). Finally, we found that a miR-29b inhibitor could mitigate the increases in osteogenic differentiation and pyroptosis and that SOCS1 showed negative regulation of osteogenic differentiation and pyroptosis in CAVD. CONCLUSION These findings suggested NLRP3 inflammasome-related genes are highly expressed in CAVD, and miR-29b reverses osteoblastic differentiation of aortic valve interstitial cells by regulating pyroptosis and inhibiting inflammation via the STAT3/SOCS1 pathway.
Collapse
Affiliation(s)
- Ming Fang
- Department of Cardiology, Hainan General Hospital, Haikou, Hainan 570311, China; Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Bin Li
- Department of Cardiology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Xinming Li
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Yudai Wang
- Department of Cardiology, Hainan General Hospital, Haikou, Hainan 570311, China
| | - Yu Zhuang
- Department of Cardiovascular surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
25
|
P2X7R Mediates the Synergistic Effect of ATP and MSU Crystals to Induce Acute Gouty Arthritis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3317307. [PMID: 36686377 PMCID: PMC9851801 DOI: 10.1155/2023/3317307] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/26/2022] [Accepted: 12/13/2022] [Indexed: 01/15/2023]
Abstract
Activation of the nod-like receptor protein 3 (NLRP3) inflammasome by monosodium urate (MSU) crystals has been identified as the molecular basis for the acute inflammatory response in gouty arthritis. However, MSU crystals alone are not sufficient to induce acute gouty arthritis (AGA). Adenosine triphosphate (ATP) is an endogenous signaling molecule involved in the NLRP3 inflammasome activation. We aimed to explore the role of ATP in MSU crystal-induced AGA development. In peripheral blood mononuclear cell-derived macrophages obtained from gout patients, we observed a synergistic effect of ATP on MSU crystal-induced IL-1β release. Furthermore, in a rat model of spontaneous gout, we demonstrated that a synergistic effect of ATP and MSU crystals, but not MSU crystals alone, is essential for triggering AGA. Mechanistically, this synergistic effect is achieved through the purinergic receptor P2X7 (P2X7R). Blockade of P2X7R prevented AGA induction in rats after local injection of MSU crystals, and carrying the mutant hP2X7R gene contributed to the inhibition of NLRP3 inflammasome activation induced by costimulation of MSU crystals and ATP in vitro. Taken together, these results support the synergistic effect of ATP on MSU crystal-induced NLRP3 inflammasome activation facilitating inflammatory episodes in AGA. In this process, P2X7R plays a key regulatory role, suggesting targeting P2X7R to be an attractive therapeutic strategy for the treatment of AGA.
Collapse
|
26
|
Shi X, Zhuang L, Zhai Z, He Y, Sun E. Polydatin protects against gouty nephropathy by inhibiting renal tubular cell pyroptosis. Int J Rheum Dis 2023; 26:116-123. [PMID: 36328529 DOI: 10.1111/1756-185x.14463] [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: 04/22/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To investigate the protective effect and mechanism of polydatin (PD) against gouty nephropathy (GN) in mice. METHODS Twenty-four mice were randomly divided into three groups: the control group (no treatment), the GN group (300 mg/kg hypoxanthine + 150 mg/kg potassium oxonate), and the GN + PD group (300 mg/kg hypoxanthine + 150 mg/kg potassium oxonate + 50 mg/kg PD). Histological changes in the kidneys and the levels of uric acid (UA), blood urea nitrogen (BUN), and serum creatinine (SCr) in the sera were measured. In addition, the expression of gasdermin D (GSDMD) protein in renal tubular epithelial cells, and the expression of NOD-like receptor protein 3 (NLRP3), GSDMD, and caspase-1 proteins in the kidney tissues were determined by immunohistochemistry, immunofluorescence, and Western blot. RESULTS In vitro, PD inhibited the expression of NLRP3, caspase-1, and GSDMD and protected the renal tubular epithelial cells from pyroptosis. In vivo, PD treatment significantly ameliorated the pathological changes in kidney tissue, and reversed the decrease of serum UA and BUN in GN model mice. The expression of NLRP3, GSDMD, and caspase-1 proteins was also decreased in the PD-treated GN mice. CONCLUSION The results suggest that PD has a protective effect on mice with GN, which may be related to the downregulation of NLRP3, GSDMD, and caspase-1 proteins and the inhibition of renal tubular epithelial cells pyroptosis.
Collapse
Affiliation(s)
- Xingliang Shi
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Institute of Clinical Immunology, Academy of Orthopedics Guangdong Province, Guangzhou, China
| | - Lili Zhuang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Institute of Clinical Immunology, Academy of Orthopedics Guangdong Province, Guangzhou, China
| | - Zeqing Zhai
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Institute of Clinical Immunology, Academy of Orthopedics Guangdong Province, Guangzhou, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Institute of Clinical Immunology, Academy of Orthopedics Guangdong Province, Guangzhou, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,Institute of Clinical Immunology, Academy of Orthopedics Guangdong Province, Guangzhou, China
| |
Collapse
|
27
|
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] [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
|
28
|
Li N, Amatjan M, He P, Wu M, Yan H, Shao X. Whole transcriptome expression profiles in kidney samples from rats with hyperuricaemic nephropathy. PLoS One 2022; 17:e0276591. [PMID: 36534664 PMCID: PMC9762607 DOI: 10.1371/journal.pone.0276591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/10/2022] [Indexed: 12/24/2022] Open
Abstract
Hyperuricaemic nephropathy (HN) is a common clinical complication of hyperuricaemia (HUA) and poses a huge threat to human health. Hence, we aimed to prospectively investigate the dysregulated genes, pathways and networks involved in HN by performing whole transcriptome sequencing using RNA sequencing. Six kidney samples from HN group (n = 3) and a control group (n = 3) were obtained to conduct RNA sequencing. To disclose the relevant signalling pathways, we conducted the analysis of differentially expressed genes (DEGs), Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. A competitive endogenous RNA (ceRNA) network was established to reveal the interactions between lncRNAs, circRNAs, mRNAs and miRNAs and investigate the potential mechanisms of HN. Ultimately, 2250 mRNAs, 306 lncRNAs, 5 circRNAs, and 70 miRNAs were determined to be significantly differentially expressed in the HN group relative to the control group. We further authenticated 8 differentially expressed (DE)-ncRNAs by quantitative real-time polymerase chain reaction, and these findings were in accordance with the sequencing results. The analysis results evidently showed that these DE-ncRNAs were significantly enriched in pathways related to inflammatory reaction. In conclusion, HUA may generate abnormal gene expression changes and regulate signalling pathways in kidney samples. Potentially related genes and pathways involved in HN were identified.
Collapse
Affiliation(s)
- Na Li
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Mukaram Amatjan
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Pengke He
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Meiwei Wu
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Hengxiu Yan
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Xiaoni Shao
- College of Pharmacy, Southwest Minzu University, Chengdu, China
- * E-mail:
| |
Collapse
|
29
|
Voronova V, Vislobokova A, Mutig K, Samsonov M, Peskov K, Sekacheva M, Materenchuk M, Bunyatyan N, Lebedeva S. Combination of immune checkpoint inhibitors with radiation therapy in cancer: A hammer breaking the wall of resistance. Front Oncol 2022; 12:1035884. [PMID: 36544712 PMCID: PMC9760959 DOI: 10.3389/fonc.2022.1035884] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022] Open
Abstract
Immuno-oncology is an emerging field in the treatment of oncological diseases, that is based on recruitment of the host immune system to attack the tumor. Radiation exposure may help to unlock the potential of the immune activating agents by enhancing the antigen release and presentation, attraction of immunocompetent cells to the inflammation site, and eliminating the tumor cells by phagocytosis, thereby leading to an overall enhancement of the immune response. Numerous preclinical studies in mouse models of glioma, murine melanoma, extracranial cancer, or colorectal cancer have contributed to determination of the optimal radiotherapy fractionation, as well as the radio- and immunotherapy sequencing strategies for maximizing the antitumor activity of the treatment regimen. At the same time, efficacy of combined radio- and immunotherapy has been actively investigated in clinical trials of metastatic melanoma, non-small-cell lung cancer and renal cell carcinoma. The present review summarizes the current advancements and challenges related to the aforementioned treatment approach.
Collapse
Affiliation(s)
- Veronika Voronova
- Department of Pharmacological Modeling, M&S Decisions LLC, Moscow, Russia
| | - Anastasia Vislobokova
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Kerim Mutig
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mikhail Samsonov
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Kirill Peskov
- Department of Pharmacological Modeling, M&S Decisions LLC, Moscow, Russia,MID3 Research Center, I.M. Sechenov First Moscow State Medical University, Moscow, Russia,Artificial Intelligence Research Center, STU Sirius, Sochi, Russia
| | - Marina Sekacheva
- World-Class Research Center “Digital biodesign and personalized healthcare”, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria Materenchuk
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Natalya Bunyatyan
- Institute of Professional Education, I.M. Sechenov First Moscow State Medical University, Moscow, Russia,Federal State Budgetary Institution “Scientific Centre for Expert Evaluation of Medicinal Products” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Svetlana Lebedeva
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia,Institute of Professional Education, I.M. Sechenov First Moscow State Medical University, Moscow, Russia,*Correspondence: Svetlana Lebedeva,
| |
Collapse
|
30
|
m(6)A methyltransferase METTL3 relieves cognitive impairment of hyperuricemia mice via inactivating MyD88/NF-κB pathway mediated NLRP3-ASC-Caspase1 inflammasome. Int Immunopharmacol 2022; 113:109375. [PMID: 36461592 DOI: 10.1016/j.intimp.2022.109375] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 10/15/2022] [Accepted: 10/16/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Recent studies have uncovered that hyperuricemia (HUA) leads to cognitive deficits, which are accompanied by neuronal damage and neuroinflammation. Here, we aim to explore the role of methyltransferase-like 3 (METTL3) in HUA-mediated neuronal apoptosis and microglial inflammation. METHODS A HUA mouse model was constructed. The spatial memory ability of the mice was assessed by the Morris water maze experiment (MWM), and neuronal apoptosis was analyzed by the TdT-mediated dUTP nick end labeling (TUNEL) assay. Besides, enzyme-linked immunosorbent assay (ELISA) was utilized to measure the contents of inflammatory factors (IL-1β, IL-6, and TNF-α) and oxidative stress markers (MDA, SOD, and CAT) in the serum of mice. In vitro, the mouse hippocampal neuron (HT22) and microglia (BV2) were treated with uric acid (UA). Flow cytometry was applied to analyze HT22 and BV2 cell apoptosis, and ELISA was conducted to observe neuroinflammation and oxidative stress. In addition, the expression of MyD88, p-NF-κB, NF-κB, NLRP3, ASC and Caspase1 was determined by Western blot. RESULTS METTL3 and miR-124-3p were down-regulated, while the MyD88-NF-κB pathway was activated in the HUA mouse model. UA treatment induced neuronal apoptosis in HT22 and stimulated microglial activation in BV2. Overexpressing METTL3 alleviated HT22 neuronal apoptosis and resisted the release of inflammatory cytokines and oxidative stress mediators in BV2 cells. METTL3 repressed MyD88-NF-κB and NLRP3-ASC-Caspase1 inflammasome. In addition, METTL3 overexpression enhanced miR-124-3p expression, while METTL3 knockdown aggravated HT22 cell apoptosis and BV2 cell overactivation. CONCLUSION METTL3 improves neuronal apoptosis and microglial activation in the HUA model by choking the MyD88/NF-κB pathway and up-regulating miR-124-3p.
Collapse
|
31
|
Liu Y, Luo D, Xu B. The combination of molecular docking and network pharmacology reveals the molecular mechanism of Danggui Niantong decoction in treating gout. Medicine (Baltimore) 2022; 101:e31535. [PMID: 36451451 PMCID: PMC9704887 DOI: 10.1097/md.0000000000031535] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Due to unhealthy diet and living habits, the incidence of gout is on the rise and has become a common disease with a high incidence. Danggui Niantong decoction (DGNTD), as a classic formula composed of 15 common herbs, has been widely used in clinical practice since ancient times to prevent and treat gout. However, the pharmacological mechanism and target of DGNTD are not clear. METHODS The potential active compounds and targets of DGNTD were obtained by traditional Chinese medicine systems pharmacology (TCMSP) database, and the differential genes of gout patients and controls were analyzed in gene expression omnibus (GEO) database. GSEA analysis of differential genes with GSEA 4.1.0 software and then the differential genes were intersected with the gout-related disease targets searched by GeneCard, CTD and OMIM disease database to obtain the final disease target. The "Traditional Chinese medicine-Active compounds-Targets" network was constructed by Cytoscape3.7.2 software. The R packet is used for enrichment analysis. The molecular docking between the active compound of DGNTD and the core target was verified by AutoDockTools software. RESULTS Two hundred eighty six and 244 targets of DGNTD-related active components and 652 targets of gout were obtained, of which 13 targets were potential targets of DGNTD in the treatment of gout. GSEA analysis showed that the differential genes were mainly involved in apoptosis, inflammatory reaction, and receptor metabolism and so on. Gene ontology (GO) functional enrichment analysis shows that DGNTD regulates many biological processes, such as the response to purine-containing compound and response to lipopolysaccharide, positive regulation of acute inflammatory response and other cellular components. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis shows that DGNTD treatment of gout is mainly related to interleukin-17 (IL-17), Toll-like receptor, rheumatoid arthritis, tumor necrosis factor (TNF) and so on. The results of molecular docking showed that the five active compounds in DGNTD had strong binding activity to core protein receptors. CONCLUSIONS The active compounds of DGNTD may achieve the purpose of treating gout by acting on the core target (CASP8, CXCL8, FOS, IL1B, IL6, JUN, PTGS2, STAT1, MMP1, TNF) to regulate cell metabolism, proliferation and apoptosis, and improve inflammatory response, which is the result of multi-component, multi-target and multi-pathway interaction. It provides an idea for the development of new combined drugs for gout.
Collapse
Affiliation(s)
- Yuan Liu
- The First Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Di Luo
- Microscopic Orthopaedic, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Bo Xu
- The First Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
- * Correspondence: Bo Xu, The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, Shandong 250000, China (e-mail: )
| |
Collapse
|
32
|
Hou W, Xia X, Li Y, Lv H, Liu J, Li X. Recent progress and perspectives on the relationship between hyperuricemia and periodontitis. Front Immunol 2022; 13:995582. [PMID: 36466813 PMCID: PMC9708725 DOI: 10.3389/fimmu.2022.995582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 11/01/2022] [Indexed: 08/31/2023] Open
Abstract
Periodontitis is one of the most prevalent diseases in oral cavity, which could not merely lead to the destruction of supporting or surrounding tooth structures but also affect the whole-body health such as the digestive and nervous systems. Epidemiological investigations suggested that in some developed countries, more than 45% or even 50% population were suffering from periodontitis. However, the prevalence increases with age remarkably and it is investigated that a high prevalence (>50%) is affecting the elderly who is over 65 years old. There is an increasing interest in the direct and indirect relationships between periodontitis and hyperuricemia. Currently, hyperuricemia has become the second major metabolic disease in modern society and the prevalence of hyperuricemia among adult males and females was 21.7% and 14.4% respectively. As an inflammatory disease associated with various systemic diseases, periodontitis may have certain connections with hyperuricemia. Partial existing research announced that hyperuricemia could act as an inhibitory factor for periodontitis, while other scholars presented that a high uric acid (UA) level was more likely to aggravate inflammatory immune response and lead to more serious tissue destruction. This article provides a detailed and comprehensive overview of the relationship underlying hyperuricemia and periodontitis in the molecular mechanisms. Given the impact of hyperuricemia, this review could provide insight into its comorbidities.
Collapse
Affiliation(s)
- Wenxue Hou
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Xiaomin Xia
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Ying Li
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Hanlin Lv
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Jie Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| | - Xue Li
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
- Dental Digital Medicine & 3D Printing Engineering Laboratory of Qingdao, Qingdao, China
- Dental Biomaterials Technology Innovation Center of Qingdao, Qingdao, China
| |
Collapse
|
33
|
The role of Nod-like receptor protein 3 inflammasome activated by ion channels in multiple diseases. Mol Cell Biochem 2022; 478:1397-1410. [PMID: 36378463 PMCID: PMC10164009 DOI: 10.1007/s11010-022-04602-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022]
Abstract
AbstractThe inflammasome is a multimeric protein complex located in the cytoplasm that is activated by many factors and subsequently promotes the release of proinflammatory factors such as interleukin (IL)-1β and IL-18, resulting in a series of inflammatory responses that ultimately lead to the occurrence of various diseases. The Nod-like receptor protein 3 (NLRP3) inflammasome is the most characteristic type and the most widely studied among many inflammasomes. Activation of the NLRP3 inflammasome is closely related to the occurrence of many diseases, such as Alzheimer's disease. At present, a large number of studies have focused on the mechanisms underlying the activation of the NLRP3 inflammasome. Plenty of articles have reported the activation of the NLRP3 inflammasome by various ions, such as K+ and Na+ reflux and Ca2+ influx. However, few articles have reviewed the effects of various ion channels on the activation of the NLRP3 inflammasome and the relationship between the diseases caused by these proteins. This article mainly summarizes the relationship between intracellular and extracellular ion activities and ion channels and the activation of the NLRP3 inflammasome. We also provide a general summary of the diseases of each system caused by NLRP3 activation. We hope that more research will provide options for the treatment of diseases driven by the NLRP3 inflammasome.
Collapse
|
34
|
Xu L, Cheng J, Lu J, Lin G, Yu Q, Li Y, Chen J, Xie J, Su Z, Zhou Q. Integrating network pharmacology and experimental validation to clarify the anti-hyperuricemia mechanism of cortex phellodendri in mice. Front Pharmacol 2022; 13:964593. [PMID: 36438835 PMCID: PMC9692208 DOI: 10.3389/fphar.2022.964593] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/24/2022] [Indexed: 08/12/2023] Open
Abstract
Hyperuricemia (HUA), a common metabolic disease, is treated as the second-largest metabolic disease after diabetes in China. Cortex Phellodendri (CP) is one of the most frequently used herbal medicines for treating gout or HUA. However, the mechanism underlying the anti-HUA effect of CP is still unrevealed. Hence, this study aimed to explore the pharmacological mechanism of CP against HUA using network pharmacology coupled with in vivo experimental validation. Active compounds and potential targets of CP, as well as the potential targets related to HUA, were retrieved from multiple open-source databases. The drug-disease overlapping targets were obtained by Venn diagram analysis and used to construct the herb-component-target (HCT), protein-protein-interaction (PPI), and component-target-pathway (CTP) networks. The functional enrichment analysis was also performed for further study. Furthermore, a HUA mouse model was induced by a combination of intraperitoneal injection of potassium oxonate (PO, 300 mg/kg) and intragastric administration of hypoxanthine (HX, 300 mg/kg) daily for 10 days. Different dosages of CP (200, 400, and 800 mg/kg) were orally given to mice 1 h after modeling. The results showed that 12 bioactive compounds and 122 drug-disease overlapping targets were obtained by matching 415 CP-related targets and 679 HUA-related targets, and berberine was one of the most important compounds with the highest degree value. The core targets of CP for treating HUA were TP53, MAPK8, MAPK3, IL-6, c-Jun, AKT1, xanthine oxidase (XOD), and ATP-binding cassette subfamily G member 2 (ABCG2). The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment results showed that the anti-HUA effect of CP mainly involved the pathways of inflammation and apoptosis, such as PI3K/Akt, TNF, MAPK, TLR, AMPK, NF-κB, and NLRP3 signaling pathways. In vivo animal experiment further confirmed the hypouricemic effect of CP in a HUA mouse model, as evidenced by significantly restored kidney histological deteriorations, and considerably decreased levels of serum uric acid (sUA), creatinine (Cre), blood urea nitrogen (BUN), and hepatic UA. Furthermore, the hypouricemic action of CP in vivo might be attributed to its suppression of XOD activity in the liver, rather than ABCG2 in the kidney. Real-time qPCR (RT-qPCR) and Western blot analysis also confirmed the key roles of the hub genes in CP against HUA. In conclusion, CP exhibited therapeutic effect against HUA via multi-compounds, multi-targets, and multi-pathways. It possessed anti-HUA and nephroprotective effects via suppressing XOD activity, and reversed the progression of renal injury by exerting anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Lieqiang Xu
- College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, China
| | - Juanjuan Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jieyi Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoshu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiuxia Yu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| |
Collapse
|
35
|
Association between Serum Uric Acid and Hypertension in a Large Cross-Section Study in a Chinese Population. J Cardiovasc Dev Dis 2022; 9:jcdd9100346. [PMID: 36286298 PMCID: PMC9604751 DOI: 10.3390/jcdd9100346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/19/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
Background: The association of serum uric acid (SUA) with hypertension has been well established in Caucasian populations. However, its association with hypertension in Chinese remained to be clarified. Methods: Consecutive patients, homogeneous in Chinese Han ethnicity, aged ≥18 years, abstracted from the database, admitted from 1 January 2010 to 31 December 2013, were included for potential analysis. The patients were grouped according to the presence or absence of hypertension. Unconditional logistic regression was performed to estimate the association between SUA and hypertension. Its possible interactions with risk factors on hypertension were also explored. Results: A total of 9587 patients were finally analyzed in the current study, where 5692 were with hypertension and 3895 were without hypertension. Per 100 μmol/L higher SUA concentration was associated with multivariable-adjusted odds ratios (95% CI) of 1.25 (1.08−1.22) in males, 1.10 (1.01−1.20) in females, and 1.19 (1.13−1.24) in total. On a categorical scale, when compared with the first quintile, the multivariable-adjusted odds ratios (95% CI) were 1.40 (1.20−1.64) for the 2nd quintile, 1.48 (1.27−1.74) for the 3rd quintile, 1.55 (1.32−1.82) for the 4th quintile, and 1.92 (1.63−2.26) for the 5th quintile, with a p for trend < 0.01. Conclusions: SUA is associated with hypertension in a dose-response manner among the Chinese hospitalized population. Management of SUA could help to the prevention and control of hypertension.
Collapse
|
36
|
Lin H, Xu Y, Zheng Y, Wu D, Ye Z, Xiao J. The association of urinary prostaglandins with uric acid in hyperuricemia patients. BMC Nephrol 2022; 23:302. [PMID: 36057582 PMCID: PMC9441060 DOI: 10.1186/s12882-022-02928-y] [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: 05/29/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose To explore the association between uric acid and urinary prostaglandins in male patients with hyperuricemia. Methods A total of 38 male patients with hyperuricemia in outpatients of Huadong Hospital from July 2018 to January 2020 were recruited. Serum uric acid (SUA), 24 h urinary uric acid excretion and other indicators were detected respectively. 10 ml urine was taken to determine prostaglandin prostaglandin D (PGD), prostaglandin E1 (PGE1), prostaglandin E2 (PGE2), 6-keto-PGF1α, thromboxane A2 (TXA2) and thromboxane B2 (TXB2). Fraction of uric acid excretion (FEua) and uric acid clearance rate (Cua) were calculated. According to the mean value of FEua and Cua, patients were divided into two groups, respectively. The independent-samples t test and the Mann–Whitney U test were applied for normally and non-normally distributed data, respectively. Results After adjusting confounding factors (age, BMI, eGFR, TG, TC, HDL and LDL), SUA was negatively correlated with urinary PGE1(r = -0.615, P = 0.009) and PGE2(r = -0.824, P < 0.001). Compared with SUA1 group (SUA < 482.6 mg/dl), SUA2 (SUA \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\ge$$\end{document}≥ 482.6 mg/dl) had lower urinary PGE1(P = 0.022) and PGE2(P = 0.019) levels. Cua was positively correlated with PGE2 (r = 0.436, P = 0.01). The correlation persisted after adjustment for age, BMI, eGFR, TG, TC, HDL and LDL by multiple linear regression analysis. In the Cua1 group (Cua < 4.869 mL /min/1.73 m2), PGE2 were lower than that in Cua2 (Cua \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\ge$$\end{document}≥ 4.869 mL /min/1.73 m2) group (P = 0.011). Conclusions In male patients with hyperuricemia, SUA was negatively correlated with urinary PGE2, Cua was positively correlated with urinary PGE2. Urinary PGE2 were significantly different between different SUA and Cua groups.
Collapse
Affiliation(s)
- Huagang Lin
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, P.R. China
| | - Ying Xu
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China
| | - Yuqi Zheng
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China
| | - Deping Wu
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, P.R. China
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, P.R. China.
| | - Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, P.R. China.
| |
Collapse
|
37
|
Guan H, Lin H, Wang X, Xu Y, Zheng Y, Zhou X, Diao X, Ye Z, Xiao J. Autophagy-dependent Na +-K +-ATPase signalling and abnormal urate reabsorption in hyperuricaemia-induced renal tubular injury. Eur J Pharmacol 2022; 932:175237. [PMID: 36063871 DOI: 10.1016/j.ejphar.2022.175237] [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: 02/24/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/03/2022]
Abstract
Increasing evidence indicates that hyperuricaemia (HUA) is not only a result of decreased renal urate excretion but also a contributor to kidney disease. Na+-K+-ATPase (NKA), which establishes the sodium gradient for urate transport in proximal tubular epithelial cells (PTECs), its impairment leads to HUA-induced nephropathy. However, the specific mechanism underlying NKA impairment-mediated renal tubular injury and increased urate reabsorption in HUA is not well understood. In this study, we investigated whether autophagy plays a key role in the NKA impairment signalling and increased urate reabsorption in HUA-induced renal tubular injury. Protein spectrum analysis of exosomes from the urine of HUA patients revealed the activation of lysosomal processes, and exosomal expression of lysosome membrane protein 2 was associated with increased serum levels and decreased renal urate excretion in patients. We demonstrated that high uric acid (UA) induced lysosome dysfunction, autophagy and inflammation in a time- and dose-dependent manner and that high UA and/or NKA α1 siRNA significantly increased mitochondrial abnormalities, such as reductions in mitochondrial respiratory complexes and cellular ATP levels, accompanied by increased apoptosis in cultured PTECs. The autophagy inhibitor hydroxychloroquine (HCQ) ameliorated NKA impairment-mediated mitochondrial dysfunction, Nod-like receptor pyrin domain-containing protein 3 (NLRP3)-interleukin-1β (IL-1β) production, and abnormal urate reabsorption in PTECs stimulated with high UA and in rats with oxonic acid (OA)-induced HUA. Our findings suggest that autophagy plays a pivotal role in NKA impairment-mediated signalling and abnormal urate reabsorption in HUA-induced renal tubular injury and that inhibition of autophagy by HCQ could be a promising treatment for HUA.
Collapse
Affiliation(s)
- Haochen Guan
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China
| | - Huagang Lin
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China
| | - Xiaojun Wang
- Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China
| | - Ying Xu
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China
| | - Yuqi Zheng
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China
| | - Xun Zhou
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China
| | - Xuehong Diao
- Department of Ultrasound, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China.
| | - Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, PR China.
| |
Collapse
|
38
|
Copur S, Demiray A, Kanbay M. Uric acid in metabolic syndrome: Does uric acid have a definitive role? Eur J Intern Med 2022; 103:4-12. [PMID: 35508444 DOI: 10.1016/j.ejim.2022.04.022] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 12/25/2022]
Abstract
Increased serum uric acid (SUA) levels are commonly seen in patients with metabolic syndrome and are widely accepted as risk factors for hypertension, gout, non-alcoholic fatty liver disease, chronic kidney disease (CKD), and cardiovascular diseases. Although some ambiguity for the exact role of uric acid (UA) in these diseases is still present, several pathophysiological mechanisms have been identified such as increased oxidative stress, inflammation, and apoptosis. Accumulating evidence in genomics enlightens genetic variabilities and some epigenetic changes that can contribute to hyperuricemia. Here we discuss the role of UA within metabolism and the consequences of asymptomatic hyperuricemia while providing newfound evidence for the associations between UA and gut microbiota and vitamin D. Increased SUA levels and beneficial effects of lowering SUA levels need to be elucidated more to understand its complicated function within different metabolic pathways and set optimal target levels for SUA for reducing risks for metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
39
|
Hu X, Liu J, Li W, Wang C, Li G, Zhou Y, Dong H. Elevated serum uric acid was associated with pre-inflammatory state and impacted the role of HDL-C on carotid atherosclerosis. Nutr Metab Cardiovasc Dis 2022; 32:1661-1669. [PMID: 35469728 DOI: 10.1016/j.numecd.2022.03.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/21/2022] [Accepted: 03/27/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND AIMS Uric acid (UA) and high-density lipoprotein cholesterol (HDL-C) are positively and negatively associated with atherosclerosis, respectively. UA and HDL-C are involved in the balance of proinflammatory and anti-inflammatory processes in atherosclerosis. However, it is still unclear whether UA affects the effect of HDL-C on atherosclerosis. METHODS AND RESULTS In this retrospective study, we enrolled 1437 patients with multiple risk factors for atherosclerosis. Patients were categorized into two groups according to their baseline UA level. Multivariate logistic regression analysis and restricted cubic spline curves were used to assess the relationship between HDL-C and carotid atherosclerosis (abnormal carotid intima-media thickness [cIMT] and carotid artery plaque) at different UA levels. Compared to patients with normouricemia, patients with hyperuricemia were older and had a more extensive history of disease and unhealthy behavior. In the normouricemia group, multivariate-adjusted odds ratios (95% CIs) for HDL-C were 0.55 (0.33-0.92) for abnormal mean cIMT, 0.59 (0.35-1.00) for abnormal maximum cIMT, and 0.53 (0.29-0.94) for the occurrence of carotid artery plaque, while the correlation between each of these three indicators with HDL-C were not significant in those with hyperuricemia. Spline regression models yielded similar results. The effect of UA on the association between HDL-C and carotid atherosclerosis remained in the subset of patients with optimal low-density lipoprotein cholesterol. CONCLUSION Elevated UA marks a pre-inflammatory state and impacts the role of HDL-C on carotid atherosclerosis.
Collapse
Affiliation(s)
- Xiangming Hu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China; Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, Guangdong, China
| | - Jieliang Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, Guangdong, China
| | - Wei Li
- Department of Cardiology, Guangdong Provincial People's Hospital Zhuhai Hospital (Zhuhai Golden Bay Center Hospital), Zhuhai 519040, Guangdong, China
| | - Chenyang Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, Guangdong, China
| | - Guang Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, Guangdong, China
| | - Yingling Zhou
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, Guangdong, China.
| | - Haojian Dong
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
40
|
Thomas JM, Huuskes BM, Sobey CG, Drummond GR, Vinh A. The IL-18/IL-18R1 signalling axis: Diagnostic and therapeutic potential in hypertension and chronic kidney disease. Pharmacol Ther 2022; 239:108191. [PMID: 35461924 DOI: 10.1016/j.pharmthera.2022.108191] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) is inherently an inflammatory condition, which ultimately results in the development of end stage renal disease or cardiovascular events. Low-grade inflammatory diseases such as hypertension and diabetes are leading causes of CKD. Declines in renal function correlate with elevated circulating pro-inflammatory cytokines in patients with these conditions. The inflammasome is an important inflammatory signalling platform that has been associated with low-grade chronic inflammatory diseases. Notably, activation and assembly of the inflammasome causes the auto cleavage of pro-caspase-1 into its active form, which then processes the pro-inflammatory cytokines pro-interleukin (IL)-1β and pro-IL-18 into their active forms. Currently, the nod-like receptor protein 3 (NLRP3) inflammasome has been implicated in the development of CKD in pre-clinical and clinical settings, and the ablation or inhibition of inflammasome components have been shown to be reno-protective in models of CKD. While clinical trials have demonstrated that neutralisation of IL-1β signalling by the drug anakinra lowers inflammation markers in haemodialysis patients, ongoing preclinical studies are showing that this ability to attenuate disease is limited in progressive models of kidney disease. These results suggest a potential predominant role for IL-18 in the development of CKD. This review will discuss the role of the inflammasome and its pro-inflammatory product IL-18 in the development of renal fibrosis and inflammation that contribute to the pathophysiology of CKD. Furthermore, we will examine the potential of the IL-18 signalling axis as an anti-inflammatory target in CKD and its usefulness as diagnostic biomarker to predict acute kidney injury.
Collapse
Affiliation(s)
- Jordyn M Thomas
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Brooke M Huuskes
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia.
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
41
|
Wang M, Lin X, Yang X, Yang Y. Research progress on related mechanisms of uric acid activating NLRP3 inflammasome in chronic kidney disease. Ren Fail 2022; 44:615-624. [PMID: 35382689 PMCID: PMC9004527 DOI: 10.1080/0886022x.2022.2036620] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
Hyperuricemia is an independent risk factor for the progression of chronic kidney disease. High levels of uric acid can lead to a series of pathological conditions, such as gout, urinary stones, inflammation, and uric acid nephropathy. There is a close relationship between uric acid and the NLRP3 inflammasome. NLRP3 inflammasome activation can cause cell damage and even death through endoplasmic reticulum stress, lysosome destruction, mitochondrial dysfunction, and the interaction between the Golgi apparatus and extracellular vesicles. In addition, the NLRP3 inflammasome acts as a molecular platform, triggering the activation of caspase-1 and the lysis of IL-1β, IL-18 and Gasdermin D (GSDMD) through different molecular mechanisms. Cleaved NT-GSDMD forms pores in the cell membrane and triggers pyrophosphorylation, thereby inducing cell death and releasing many intracellular proinflammatory molecules. In recent years, studies have found that hyperuricemia or uric acid crystals can activate NLRP3 inflammasomes, and the activation of NLRP3 inflammasomes plays an important role in kidney disease. This article reviews the possible pathophysiological mechanisms by which uric acid activates inflammasomes and induces kidney damage at the cellular and molecular levels.
Collapse
Affiliation(s)
- Miao Wang
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Xin Lin
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Xiaoming Yang
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Yanlang Yang
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
42
|
Nutmakul T. A review on benefits of quercetin in hyperuricemia and gouty arthritis. Saudi Pharm J 2022; 30:918-926. [PMID: 35903522 PMCID: PMC9315272 DOI: 10.1016/j.jsps.2022.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Hyperuricemia becomes a public health problem worldwide. It is not only a major risk factor for gout but also associated with the development of life-threatening diseases such as chronic kidney disease and cardiovascular diseases. Although there are several available therapeutic drugs, some serious adverse effects and contraindications are concerned. These drive the search for an alternative therapy that is effective and safe. Quercetin is of particular interesting since it has been reported numerous pharmacological activities, especially anti-hyperuricemia, antioxidant, anti-inflammation and amelioration of metabolic syndromes and cardiovascular diseases which are comorbidities of hyperuricemia and gout. In addition, quercetin has been widely used as a health supplement for many diseases however, the use for hyperuricemia and gout has not been indicated. Therefore, this review aims to gather and summarize published data regarding the efficacy in preclinical and clinical studies along with possible mechanism of action, and safety aspect of quercetin in order to support the use of quercetin as a dietary supplement for prevention and management of hyperuricemia and gouty arthritis and/or use as alternative or combination therapy to minimize the side effects of the conventional drugs.
Collapse
|
43
|
He S, Xiong Q, Li L, Lin X, Zhao J, Guo X, He Y, Liang W, Ying C, Zuo X. Increased risk of modality failure with higher serum uric acid level in continuous ambulatory peritoneal dialysis patients: a prospective cohort study. Ren Fail 2022; 44:272-281. [PMID: 35172675 PMCID: PMC8856069 DOI: 10.1080/0886022x.2022.2035762] [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] [Indexed: 10/29/2022] Open
Abstract
BACKGROUND Peritoneal dialysis (PD) is one of the most important kidney replacement therapies for patients with end-stage kidney disease (ESKD). PD technique failure can lead to an escalated cost and increased infectious and cardiovascular risk, up and including to death. The accumulation of uric acid (UA) was associated with adverse outcomes in ESKD patients. However, the relationship between serum UA and technique failure is little explored. METHODS Here, a total of 266 continuous ambulatory peritoneal dialysis (CAPD) patients (age, 41.8 ± 12.6 years; 125 males) were enrolled and followed up for 31.7 months. Serum UA levels were examined at baseline and each visit. Subjects were divided into three groups according to their baseline serum UA concentrations. Multivariable Cox regression models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs) of PD technique failure. RESULTS The level of serum UA increased gradually as time prolonged. During the follow-up period, 77 (28.9%) patients occurred PD technique failure, of which 56 (21.1%) transferred to hemodialysis (HD) and 21 (7.9%) died. Compared to the lowest UA tertile, after adjusting for potential confounders, HRs of technique failure in tertile 2 and tertile 3 were 1.82 (95% CI: 0.95-3.49) and 2.03 (95% CI: 1.05-3.92), respectively, and p for trend was 0.043. Adjusted HRs of all-cause technique failure, transferring to HD and mortality with each 1 mg/dL increase in serum UA were 1.20 (95% CI: 1.03-1.40, p = 0.019), 1.22 (95% CI: 1.01-1.48, p = 0.039), and 1.25 (95% CI: 0.94-1.67, p = 0.128), respectively. CONCLUSION Higher serum UA level predicted higher risk of technique failure in CAPD patients.
Collapse
Affiliation(s)
- Shuiqing He
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qianqian Xiong
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Li
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuechun Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Zhao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaolei Guo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuqin He
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wangqun Liang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chenjiang Ying
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuezhi Zuo
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
44
|
Hosoya T, Uchida S, Shibata S, Tomioka NH, Matsumoto K, Hosoyamada M. Xanthine Oxidoreductase Inhibitors Suppress the Onset of Exercise-Induced AKI in High HPRT Activity Urat1- Uox Double Knockout Mice. J Am Soc Nephrol 2022; 33:326-341. [PMID: 34799437 PMCID: PMC8819989 DOI: 10.1681/asn.2021050616] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Hereditary renal hypouricemia type 1 (RHUC1) is caused by URAT1/SLC22A12 dysfunction, resulting in urolithiasis and exercise-induced AKI (EIAKI). However, because there is no useful experimental RHUC1 animal model, the precise pathophysiologic mechanisms underlying EIAKI have yet to be elucidated. We established a high HPRT activity Urat1-Uox double knockout (DKO) mouse as a novel RHUC1 animal model for investigating the cause of EIAKI and the potential therapeutic effect of xanthine oxidoreductase inhibitors (XOIs). METHODS The novel Urat1-Uox DKO mice were used in a forced swimming test as loading exercise to explore the onset mechanism of EIAKI and evaluate related purine metabolism and renal injury parameters. RESULTS Urat1-Uox DKO mice had uricosuric effects and elevated levels of plasma creatinine and BUN as renal injury markers, and decreased creatinine clearance observed in a forced swimming test. In addition, Urat1-Uox DKO mice had increased NLRP3 inflammasome activity and downregulated levels of Na+-K+-ATPase protein in the kidney, as Western blot analysis showed. Finally, we demonstrated that topiroxostat and allopurinol, XOIs, improved renal injury and functional parameters of EIAKI. CONCLUSIONS Urat1-Uox DKO mice are a useful experimental animal model for human RHUC1. The pathogenic mechanism of EIAKI was found to be due to increased levels of IL-1β via NLRP3 inflammasome signaling and Na+-K+-ATPase dysfunction associated with excessive urinary urate excretion. In addition, XOIs appear to be a promising therapeutic agent for the treatment of EIAKI.
Collapse
Affiliation(s)
- Takuji Hosoya
- Department of Human Physiology and Pathology, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan,Biological Research Department, Research Institute, Fuji Yakuhin Co., Ltd., Saitama, Japan
| | - Shunya Uchida
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Naoko H. Tomioka
- Department of Human Physiology and Pathology, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Koji Matsumoto
- Biological Research Department, Research Institute, Fuji Yakuhin Co., Ltd., Saitama, Japan
| | - Makoto Hosoyamada
- Department of Human Physiology and Pathology, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| |
Collapse
|
45
|
Hypothetical Mechanism of Exercise-Induced Acute Kidney Injury Associated with Renal Hypouricemia. Biomedicines 2021; 9:biomedicines9121847. [PMID: 34944661 PMCID: PMC8698656 DOI: 10.3390/biomedicines9121847] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 11/17/2022] Open
Abstract
Renal hypouricemia (RHUC) is a hereditary disease that presents with increased renal urate clearance and hypouricemia due to genetic mutations in the urate transporter URAT1 or GLUT9 that reabsorbs urates in the renal proximal tubule. Exercise-induced acute kidney injury (EIAKI) is known to be a complication of renal hypouricemia. In the skeletal muscle of RHUC patients during exhaustive exercise, the decreased release of endothelial-derived hyperpolarization factor (EDHF) due to hypouricemia might cause the disturbance of exercise hyperemia, which might increase post-exercise urinary urate excretion. In the kidneys of RHUC patients after exhaustive exercise, an intraluminal high concentration of urates in the proximal straight tubule and/or thick ascending limb of Henle’s loop might stimulate the luminal Toll-like receptor 4–myeloid differentiation factor 88–phosphoinositide 3-kinase–mammalian target of rapamycin (luminal TLR4–MyD88–PI3K–mTOR) pathway to activate the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome and may release interleukin-1β (IL-1β), which might cause the symptoms of EIAKI.
Collapse
|
46
|
Liu ZQ, Huang ZW, Kang SL, Hu CC, Chen F, He F, Lin Z, Yang F, Hu ZJ. Serum Uric Acid and Cardiovascular or All-Cause Mortality in Peritoneal Dialysis Patients: A Systematic Review and Meta-Analysis. Front Cardiovasc Med 2021; 8:751182. [PMID: 34805305 PMCID: PMC8597842 DOI: 10.3389/fcvm.2021.751182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/11/2021] [Indexed: 11/26/2022] Open
Abstract
Background: Studies have shown inconsistent associations between serum uric acid (SUA) levels and mortality in peritoneal dialysis (PD) patients. We conducted this meta-analysis to determine whether SUA levels were associated with cardiovascular or all-cause mortality in PD patients. Methods: PubMed, Embase, Web of Science, the Cochrane Library, CNKI, VIP, Wanfang Database, and trial registry databases were systematically searched up to April 11, 2021. Cohort studies of SUA levels and cardiovascular or all-cause mortality in PD patients were obtained. Random effect models were used to calculate the pooled adjusted hazard ratio (HR) and corresponding 95% confidence interval (CI). Sensitivity analyses were conducted to assess the robustness of the pooled results. Subgroup analyses and meta-regression analyses were performed to explore the sources of heterogeneity. Funnel plots, Begg's tests, and Egger's tests were conducted to evaluate potential publication bias. The GRADE approach was used to rate the certainty of evidence. This study was registered with PROSPERO, CRD42021268739. Results: Seven studies covering 18,113 PD patients were included. Compared with the middle SUA levels, high SUA levels increased the risk of all-cause mortality (HR = 1.74, 95%CI: 1.26–2.40, I2 = 34.8%, τ2 = 0.03), low SUA levels were not statistically significant with the risk of all-cause or cardiovascular mortality (HR = 1.04, 95%CI: 0.84–1.29, I2 = 43.8%, τ2 = 0.03; HR = 0.89, 95%CI: 0.65–1.23, I2 = 36.3%, τ2 = 0.04; respectively). Compared with the low SUA levels, high SUA levels were not statistically associated with an increased risk of all-cause or cardiovascular mortality (HR = 1.19, 95%CI: 0.59–2.40, I2 = 88.2%, τ2 = 0.44; HR = 1.22, 95%CI: 0.39–3.85, I2 = 89.3%, τ2 = 0.92; respectively). Conclusion: Compared with middle SUA levels, high SUA levels are associated with an increased risk of all-cause mortality in PD patients. SUA levels may not be associated with cardiovascular mortality. More high-level studies, especially randomized controlled trials, are needed to determine the association between SUA levels and cardiovascular or all-cause mortality in PD patients. Systematic Review Registration:https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021268739, identifier: CRD42021268739.
Collapse
Affiliation(s)
- Zhi-Qiang Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Zhi-Wen Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Shu-Ling Kang
- Fuzhou Center for Disease Control and Prevention, Fuzhou, China.,Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Chan-Chan Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Fa Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Fei He
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Zheng Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Feng Yang
- Department of Nephrology, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, China
| | - Zhi-Jian Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| |
Collapse
|
47
|
Goldberg A, Garcia-Arroyo F, Sasai F, Rodriguez-Iturbe B, Sanchez-Lozada LG, Lanaspa MA, Johnson RJ. Mini Review: Reappraisal of Uric Acid in Chronic Kidney Disease. Am J Nephrol 2021; 52:837-844. [PMID: 34673651 DOI: 10.1159/000519491] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022]
Abstract
Hyperuricemia predicts the development of chronic kidney disease (CKD) and metabolic complications, but whether it has a causal role has been controversial. This is especially true given the 2 recently conducted randomized controlled trials that failed to show a benefit of lowering uric acid in type 1 diabetes-associated CKD and subjects with stage 3-4 CKD. While these studies suggest that use of urate-lowering drugs in unselected patients is unlikely to slow the progression of CKD, there are subsets of subjects with CKD where reducing uric acid synthesis may be beneficial. This may be the case in patients with gout, hyperuricemia (especially associated with increased production), and urate crystalluria. Here, we discuss the evidence and propose that future clinical trials targeting these specific subgroups should be performed.
Collapse
Affiliation(s)
- Avi Goldberg
- Clalit Health Services, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Fumihiko Sasai
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | | | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain VA Medical Center, Aurora, Colorado, USA
| |
Collapse
|
48
|
Hyperuricemia and Progression of Chronic Kidney Disease: A Review from Physiology and Pathogenesis to the Role of Urate-Lowering Therapy. Diagnostics (Basel) 2021; 11:diagnostics11091674. [PMID: 34574015 PMCID: PMC8466342 DOI: 10.3390/diagnostics11091674] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/19/2022] Open
Abstract
The relationship between hyperuricemia, gout, and renal disease has been investigated for several years. From the beginning, kidney disease has been considered a complication of gout; however, the viewpoints changed, claiming that hypertension and elevated uric acid (UA) levels are caused by decreased urate excretion in patients with renal impairment. To date, several examples of evidence support the role of hyperuricemia in cardiovascular or renal diseases. Several mechanisms have been identified that explain the relationship between hyperuricemia and chronic kidney disease, including the crystal effect, renin-angiotensin-aldosterone system activation, nitric oxide synthesis inhibition, and intracellular oxidative stress stimulation, and urate-lowering therapy (ULT) has been proven to reduce renal disease progression in the past few years. In this comprehensive review, the source and physiology of UA are introduced, and the mechanisms that explain the reciprocal relationship between hyperuricemia and kidney disease are reviewed. Lastly, current evidence supporting the use of ULT to postpone renal disease progression in patients with hyperuricemia and gout are summarized.
Collapse
|
49
|
Clavijo-Cornejo D, Hernández-González O, Gutierrez M. The current role of NLRP3 inflammasome polymorphism in gout susceptibility. Int J Rheum Dis 2021; 24:1257-1265. [PMID: 34390315 DOI: 10.1111/1756-185x.14205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The NLR family pyrin domain containing 3 (NLRP3) signaling pathway has an important role in inflammation mediated by monosodium urate crystals in gout, and the characterization of single nucleotide polymorphisms (SNPs) have helped to recognize disease susceptibility. OBJECTIVE The aim of this review is to provide an overview of the potential role of the inflammasome gene SNPs as a susceptibility factor for gout, discussing the current evidence available. METHODS This review analyzes the relevant literature in the field of inflammasome SNPs and gout published in the last 10 years. The systematic research was performed in 16 articles, including both the SNPs associated and those not associated with gout, with the goal to have a complete overview. RESULTS Sixty-nine SNPs from 10 different genes have been reported in the literature. Of these, 13 SNPs present association with gout susceptibility in different populations, while 56 have been established as not being associated with the disease. CONCLUSIONS This review is a summary of the potential role of inflammasome gene SNPs and their association with gout risk, all of them related with NLRP3 inflammasome signaling, suggesting these polymorphisms are susceptibility candidates and genetic markers for gout. From the 69 SNPs analyzed in the literature, 13 of them have been associated with gout as follows: NLRP3 (rs3806268 and rs10754558), CARD8 (rs2043211), TLR4 (rs2149356), CD14 (rs2569190), IL-1β (rs1143623), P2RX7 (rs2230911, rs1653624, rs7958316 and rs17525809) and PPARGC1B (rs45520937, rs10491360 and rs7712296) in different populations.
Collapse
Affiliation(s)
- Denise Clavijo-Cornejo
- Division of Musculoskeletal and Rheumatic Diseases, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | | | - Marwin Gutierrez
- Division of Musculoskeletal and Rheumatic Diseases, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| |
Collapse
|
50
|
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
|