1
|
Liu M, Shi S, Guo Y, Lv M, Feng X, Liu T, Yang H, Zhang Z. Potential Biomarkers for Neurobrucellosis Diagnosis and Treatment Based on Cerebrospinal Fluid Metabolomics Analysis. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e9946. [PMID: 39601633 DOI: 10.1002/rcm.9946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Zhenzhu Tongluo pills (ZZTL) have been utilized for the treatment of neurobrucellosis (NB) in the Inner Mongolia region of China. However, the specific mechanism underlying the neuroprotective effects of ZZTL in NB remains insufficiently explored. Therefore, this study aimed to analyze the metabolite profiles across different groups to identify potential biomarkers for the diagnosis and treatment of NB. METHODS LC-MS analysis was used to screen differential metabolites in cerebrospinal fluid (CSF) samples from control subjects, NB patients, and NB patients undergoing ZZTL therapy. RESULTS We identified 225 common metabolites across two comparison groups (NB vs. control group and NB + ZZTL vs. NB group). Among these metabolites, 138 were downregulated, and 87 were upregulated in the NB group compared to the control group, while the levels of these metabolites were reversed by ZZTL therapy. Creatinine exhibited the highest VIP score in both comparison groups, with elevated levels observed in CSF samples from NB patients compared to controls; however, its levels were reduced following ZZTL therapy. Additionally, hypoxanthine and uric acids levels were also increased in CSF samples from NB patients, indicating dysregulation of purine metabolism in NB; however, these changes were reversed by ZZTL. Furthermore, pantothenic acid (vitamin B5) and niacinamide (vitamin B3) levels were decreased in CSF samples from NB patients, suggesting a potential link between NB and vitamin B3 and B5 deficiency; however, these changes were reversed by ZZTL. CONCLUSION Collectively, CSF metabolomics may effectively differentiate NB patients from control subjects, providing valuable insights for NB diagnosis and treatment.
Collapse
Affiliation(s)
- Meiling Liu
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Shujun Shi
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yuzhou Guo
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Menghan Lv
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xuan Feng
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Ting Liu
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hao Yang
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital & Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhelin Zhang
- Department of Neurology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
2
|
Du L, Zong Y, Li H, Wang Q, Xie L, Yang B, Pang Y, Zhang C, Zhong Z, Gao J. Hyperuricemia and its related diseases: mechanisms and advances in therapy. Signal Transduct Target Ther 2024; 9:212. [PMID: 39191722 DOI: 10.1038/s41392-024-01916-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/08/2024] [Accepted: 06/27/2024] [Indexed: 08/29/2024] Open
Abstract
Hyperuricemia, characterized by elevated levels of serum uric acid (SUA), is linked to a spectrum of commodities such as gout, cardiovascular diseases, renal disorders, metabolic syndrome, and diabetes, etc. Significantly impairing the quality of life for those affected, the prevalence of hyperuricemia is an upward trend globally, especially in most developed countries. UA possesses a multifaceted role, such as antioxidant, pro-oxidative, pro-inflammatory, nitric oxide modulating, anti-aging, and immune effects, which are significant in both physiological and pathological contexts. The equilibrium of circulating urate levels hinges on the interplay between production and excretion, a delicate balance orchestrated by urate transporter functions across various epithelial tissues and cell types. While existing research has identified hyperuricemia involvement in numerous biological processes and signaling pathways, the precise mechanisms connecting elevated UA levels to disease etiology remain to be fully elucidated. In addition, the influence of genetic susceptibilities and environmental determinants on hyperuricemia calls for a detailed and nuanced examination. This review compiles data from global epidemiological studies and clinical practices, exploring the physiological processes and the genetic foundations of urate transporters in depth. Furthermore, we uncover the complex mechanisms by which the UA induced inflammation influences metabolic processes in individuals with hyperuricemia and the association with its relative disease, offering a foundation for innovative therapeutic approaches and advanced pharmacological strategies.
Collapse
Grants
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
Collapse
Affiliation(s)
- Lin Du
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Haorui Li
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Qiyue Wang
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Lei Xie
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Bo Yang
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Zhigang Zhong
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China.
| | - Junjie Gao
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China.
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
3
|
Liu J, Lin C, Wu M, Wang Y, Chen S, Yang T, Xie C, Kong Y, Wu W, Wang J, Ma X, Teng C. Co-delivery of indomethacin and uricase as a new strategy for inflammatory diseases associated with high uric acid. Drug Deliv Transl Res 2024; 14:1820-1838. [PMID: 38127247 DOI: 10.1007/s13346-023-01487-5] [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] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
Uric acid is the final metabolite in humans. High level of uric acid chronically induces urate deposition, aggravates kidney damage, and concomitantly causes an increase in inflammatory factors. Alleviating acute inflammation and decreasing uric acid levels are the key points in the treatment of inflammatory diseases associated with high uric acid. However, a drug delivery system that combines anti-inflammatory and uric acid reduction functions at the same time remains a challenge to be settled. Here, we designed a nanocrystal-based co-delivery platform, IND Nplex, characterized by loading of indomethacin (IND) and uricase. Compared with free IND or uricase, IND Nplex possessed a better anti-inflammatory effect by restraining the release of inflammation-related factors in vitro. In addition, pharmacokinetic and biodistribution studies revealed that IND Nplex significantly prolonged the retention time in vivo and was more concentrated in the kidney. In acute gouty arthritis model rats, IND Nplex markedly relieved ankle joint swelling and mitigated synovial inflammation. In acute kidney injury model rats, IND Nplex indicated better biocompatibility and significant amelioration of renal fibrosis. Moreover, IND Nplex showed the effect of anti-inflammatory and improved renal function via determination of inflammatory factors and biochemical markers in the serum and kidney. In conclusion, these results indicate that IND Nplex exerts anti-inflammatory activity and uric acid-lowering effect and could become a promising candidate for the treatment of uric acid-related diseases.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Chenshi Lin
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Man Wu
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Yingjie Wang
- Center for Translational Imaging, Northeastern University, 360 Huntington Ave., Boston, MA, 02115, USA
| | - Shenyu Chen
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Taiwang Yang
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Chenlu Xie
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Yue Kong
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Wenliang Wu
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Jiaping Wang
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Xiaonan Ma
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 210009, China.
| | - Chao Teng
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
4
|
Zheng Y, Chen Z, Yang J, Zheng J, Shui X, Yan Y, Huang S, Liang Z, Lei W, He Y. The Role of Hyperuricemia in Cardiac Diseases: Evidence, Controversies, and Therapeutic Strategies. Biomolecules 2024; 14:753. [PMID: 39062467 PMCID: PMC11274514 DOI: 10.3390/biom14070753] [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: 05/13/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024] Open
Abstract
Hyperuricemia (HUA) may lead to myocardial cell damage, thereby promoting the occurrence and adverse outcomes of heart diseases. In this review, we discuss the latest clinical research progress, and explore the impact of HUA on myocardial damage-related diseases such as myocardial infarction, arrhythmias, and heart failure. We also combined recent findings from basic research to analyze potential mechanisms linking HUA with myocardial injury. In different pathological models (such as direct action of high uric acid on myocardial cells or combined with myocardial ischemia-reperfusion model), HUA may cause damage by activating the NOD-like receptor protein 3 inflammasome-induced inflammatory response, interfering with cardiac cell energy metabolism, affecting antioxidant defense systems, and stimulating reactive oxygen species production to enhance the oxidative stress response, ultimately resulting in decreased cardiac function. Additionally, we discuss the impact of lowering uric acid intervention therapy and potential safety issues that may arise. However, as the mechanism underlying HUA-induced myocardial injury is poorly defined, further research is warranted to aid in the development novel therapeutic strategies for HUA-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yue Zheng
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhirui Chen
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Jinya Yang
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, USA;
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China;
| | - Yiguang Yan
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (S.H.); (Z.L.)
| | - Shian Huang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (S.H.); (Z.L.)
| | - Zheng Liang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (S.H.); (Z.L.)
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
- Precision Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| |
Collapse
|
5
|
Chaudhary A, He Z, Atwood DJ, Miyazaki M, Oto OA, Davidoff A, Edelstein CL. Raising serum uric acid with a uricase inhibitor worsens PKD in rat and mouse models. Am J Physiol Renal Physiol 2024; 326:F1004-F1015. [PMID: 38634129 DOI: 10.1152/ajprenal.00372.2023] [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: 12/05/2023] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Humans are predisposed to gout because they lack uricase that converts uric acid to allantoin. Rodents have uricase, resulting in low basal serum uric acid. A uricase inhibitor raises serum uric acid in rodents. There were two aims of the study in polycystic kidney disease (PKD): 1) to determine whether increasing serum uric acid with the uricase inhibitor, oxonic acid, resulted in faster cyst growth and 2) to determine whether treatment with the xanthine oxidase inhibitor, oxypurinol, reduced the cyst growth caused by oxonic acid. Orthologous models of human PKD were used: PCK rats, a polycystic kidney and hepatic disease 1 (Pkhd1) gene model of autosomal recessive PKD (ARPKD) and Pkd1RC/RC mice, a hypomorphic Pkd1 gene model. In PCK rats and Pkd1RC/RC mice, oxonic acid resulted in a significant increase in serum uric acid, kidney weight, and cyst index. Mechanisms of increased cyst growth that were investigated were proinflammatory cytokines, the inflammasome, and crystal deposition in the kidney. Oxonic acid resulted in an increase in proinflammatory cytokines in the serum and kidney in Pkd1RC/RC mice. Oxonic acid did not cause activation of the inflammasome or uric acid crystal deposition in the kidney. In Pkd1RC/RC male and female mice analyzed together, oxypurinol decreased the oxonic acid-induced increase in cyst index. In summary, increasing serum uric acid by inhibiting uricase with oxonic acid results in an increase in kidney weight and cyst index in PCK rats and Pkd1RC/RC mice. The effect is independent of inflammasome activation or crystal deposition in the kidney.NEW & NOTEWORTHY This is the first reported study of uric acid measurements and xanthine oxidase inhibition in polycystic kidney disease (PKD) rodents. Raising serum uric acid with a uricase inhibitor resulted in increased kidney weight and cyst index in Pkd1RC/RC mice and PCK rats, elevated levels of proinflammatory cytokines in the serum and kidney in Pkd1RC/RC mice, and no uric acid crystal deposition or activation of the caspase-1 inflammasome in the kidney.
Collapse
Affiliation(s)
- Anjana Chaudhary
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Zhibin He
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Daniel J Atwood
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Makoto Miyazaki
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Ozgur A Oto
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | | | - Charles L Edelstein
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
6
|
Laudisio A, Dorizzi A, Villeggia F, Latino F, Filippucci D, Zuccalà G. Association of serum uric acid with functional disability in older subjects: a population-based study. Aging Clin Exp Res 2024; 36:96. [PMID: 38630352 PMCID: PMC11023951 DOI: 10.1007/s40520-024-02746-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND The role of serum uric acid (SUA) in the development of adverse health outcomes in advanced age is still uncertain. AIMS The aim of the study was to assess the association of disability with SUA levels in older community-dwelling subjects. METHODS We assessed the association of disability with SUA in all 351 inhabitants of Tuscania (Italy) aged 75+. Functional ability was estimated using the instrumental activities of daily living (IADLs). RESULTS In logistic regression, increasing SUA levels were associated with disability (OR = 1.22; 95%CI = 1.01-1.48; P = .036), after adjusting. The association was independent of both gender and age (P for interaction > 0.050). SUA levels above 5.5 mg/dL best predicted disability. CONCLUSIONS In older subjects, SUA levels are associated with disability; the cut off level above 5.5 mg/dL might be adopted in pharmacological trials aiming at reducing the incidence and progression of disability by reducing SUA, and for identifying subjects at increased risk of disability.
Collapse
Affiliation(s)
- Alice Laudisio
- Department of Medicine, Research Unit of Geriatrics, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, Roma, 21 - 00128, Italy.
- Operative Research Unit of Geriatrics, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, Roma, 200 - 00128, Italy.
| | - Agnese Dorizzi
- Department of Medicine, Research Unit of Geriatrics, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, Roma, 21 - 00128, Italy
| | - Fabio Villeggia
- Department of Medicine, Research Unit of Geriatrics, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, Roma, 21 - 00128, Italy
| | - Francesca Latino
- Department of Medicine, Research Unit of Geriatrics, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, Roma, 21 - 00128, Italy
| | - Daniele Filippucci
- Department of Medicine, Research Unit of Geriatrics, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, Roma, 21 - 00128, Italy
| | - Giuseppe Zuccalà
- Department of Geriatric and Orthopaedic Sciences, Catholic University of Medicine, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, Rome, 00168, Italy
| |
Collapse
|
7
|
Rao B, Xie D, Deng Y, Ye J, Zeng X, Lin A, Chen J, Huang D, Xie C, Chen C, Luo Y, Lu X, Wang X, Lu J. Robust positive association between serum urate and the risk of chronic obstructive pulmonary disease: hospital-based cohort and Mendelian randomisation study. BMJ Open Respir Res 2024; 11:e002203. [PMID: 38479817 PMCID: PMC10941131 DOI: 10.1136/bmjresp-2023-002203] [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: 11/22/2023] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) and hyperuricaemia are both characterised by systemic inflammation. Preventing chronic diseases among the population with common metabolic abnormality is an effective strategy. However, the association of hyperuricaemia with the higher incidence and risk of COPD remains controversial. Therefore, replicated researches in populations with distinct characteristics or demographics are compellingly warranted. METHODS This cohort study adopted a design of ambispective hospital-based cohort. We used propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) to minimise the effects of potential confounding factors. A Cox regression model and restricted cubic spline (RCS) model were applied further to assess the effect of serum urate on the risk of developing COPD. Finally, we conducted a two-sample Mendelian randomisation (MR) analysis to explore evidence of causal association. RESULTS There is a higher incidence in the population with hyperuricaemia compared with the population with normal serum urate (22.29/1000 person-years vs 8.89/1000 person-years, p=0.009). This result is robust after performing PSM (p=0.013) and IPTW (p<0.001). The Cox model confirms that hyperuricaemia is associated with higher risk of developing COPD (adjusted HR=3.35 and 95% CI=1.61 to 6.96). Moreover, RCS shows that the risk of developing COPD rapidly increases with the concentration of serum urate when it is higher than the reference (420 µmol/L). Finally, in MR analysis, the inverse variance weighted method evidences that a significant causal effect of serum urate on COPD (OR=1.153, 95% CI=1.034 to 1.289) is likely to be true. The finding of MR is robust in the repeated analysis using different methods and sensitivity analysis. CONCLUSIONS Our study provides convincing evidence suggesting a robust positive association between serum urate and the risk of developing COPD, and indicates that the population with hyperuricaemia is at high risk of COPD in the Chinese population who seek medical advice or treatment in the hospital.
Collapse
Affiliation(s)
- Boqi Rao
- Emergency Department, The Second Affiliated Hospital, The State Key Lab of Respiratory Disease, Institute of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dongming Xie
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yibin Deng
- Centre for Medical Laboratory Science, the Afliated Hospital of Youjiang Medical University for Nationalities, Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi, Baise, Guangxi, China
| | - Junyi Ye
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaobin Zeng
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ao Lin
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinbin Chen
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dongsheng Huang
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Chenli Xie
- Department of Respiratory and Critical Care Medicine, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong, China
- Dongguan Key Laboratory of Precision, Dongguan, Guangdong, China
| | - Cuiyi Chen
- Department of Respiratory and Critical Care Medicine, Songshan Lake Central Hospital of Dongguan, Dongguan, Guangdong, China
| | - Yixuan Luo
- Guangzhou Panyu District Central Hospital, Guangzhou, Guangdong, China
| | - Xiaoxiao Lu
- Department of English and American Studies, Ludwig Maximilian University of Munich Faculty of Languages and Literatures, Munich, Germany
| | - Xinhua Wang
- Institute of Basic Medicine, Institute of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jiachun Lu
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Yanai H, Adachi H, Hakoshima M, Iida S, Katsuyama H. A Possible Therapeutic Application of the Selective Inhibitor of Urate Transporter 1, Dotinurad, for Metabolic Syndrome, Chronic Kidney Disease, and Cardiovascular Disease. Cells 2024; 13:450. [PMID: 38474414 DOI: 10.3390/cells13050450] [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: 12/21/2023] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
The reabsorption of uric acid (UA) is mainly mediated by urate transporter 1 (URAT1) and glucose transporter 9 (GLUT9) in the kidneys. Dotinurad inhibits URAT1 but does not inhibit other UA transporters, such as GLUT9, ATP-binding cassette transporter G2 (ABCG2), and organic anion transporter 1/3 (OAT1/3). We found that dotinurad ameliorated the metabolic parameters and renal function in hyperuricemic patients. We consider the significance of the highly selective inhibition of URAT1 by dotinurad for metabolic syndrome, chronic kidney disease (CKD), and cardiovascular disease (CVD). The selective inhibition of URAT1 by dotinurad increases urinary UA in the proximal tubules, and this un-reabsorbed UA may compete with urinary glucose for GLUT9, reducing glucose reabsorption. The inhibition by dotinurad of UA entry via URAT1 into the liver and adipose tissues increased energy expenditure and decreased lipid synthesis and inflammation in rats. Such effects may improve metabolic parameters. CKD patients accumulate uremic toxins, including indoxyl sulfate (IS), in the body. ABCG2 regulates the renal and intestinal excretion of IS, which strongly affects CKD. OAT1/3 inhibitors suppress IS uptake into the kidneys, thereby increasing plasma IS, which produces oxidative stress and induces vascular endothelial dysfunction in CKD patients. The highly selective inhibition of URAT1 by dotinurad may be beneficial for metabolic syndrome, CKD, and CVD.
Collapse
Affiliation(s)
- Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Ichikawa 272-8516, Chiba, Japan
| | - Hiroki Adachi
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Ichikawa 272-8516, Chiba, Japan
| | - Mariko Hakoshima
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Ichikawa 272-8516, Chiba, Japan
| | - Sakura Iida
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Ichikawa 272-8516, Chiba, Japan
| | - Hisayuki Katsuyama
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Ichikawa 272-8516, Chiba, Japan
| |
Collapse
|
9
|
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
|
10
|
Notsu T, Kurata Y, Ninomiya H, Taufiq F, Komatsu K, Miake J, Sawano T, Tsuneto M, Shirayoshi Y, Hisatome I. Inhibition of the uric acid efflux transporter ABCG2 enhances stimulating effect of soluble uric acid on IL-1β production in murine macrophage-like J774.1 cells. Hypertens Res 2023; 46:2368-2377. [PMID: 37592041 DOI: 10.1038/s41440-023-01391-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/12/2023] [Accepted: 07/08/2023] [Indexed: 08/19/2023]
Abstract
Soluble uric acid (UA) absorbed by cells through UA transporters (UATs) accumulates intracellularly, activates the NLRP3 inflammasome and thereby increases IL-1β secretion. ABCG2 transporter excludes intracellular UA. However, it remains unknown whether ABCG2 inhibition leads to intracellular accumulation of UA and increases IL-1β production. In this study, we examined whether genetic and pharmacological inhibition of ABCG2 could increase IL-1β production in mouse macrophage-like J774.1 cells especially under hyperuricemic conditions. We determined mRNA and protein levels of pro-IL-1β, mature IL-1β, caspase-1 and several UATs in culture supernatants and lysates of J774.1 cells with or without soluble UA pretreatment. Knockdown experiments using an shRNA against ABCG2 and pharmacological experiments with an ABCG2 inhibitor were conducted. Extracellularly applied soluble UA increased protein levels of pro-IL-1β, mature IL-1β and caspase-1 in the culture supernatant from lipopolysaccharide (LPS)-primed and monosodium urate crystal (MSU)-stimulated J774.1 cells. J774.1 cells expressed UATs of ABCG2, GLUT9 and MRP4, and shRNA knockdown of ABCG2 increased protein levels of pro-IL-1β and mature IL-1β in the culture supernatant. Soluble UA increased mRNA and protein levels of ABCG2 in J774.1 cells without either LPS or MSU treatment. An ABCG2 inhibitor, febuxostat, but not a urate reabsorption inhibitor, dotinurad, enhanced IL-1β production in cells pretreated with soluble UA. In conclusion, genetic and pharmacological inhibition of ABCG2 enhanced IL-1β production especially under hyperuricemic conditions by increasing intracellularly accumulated soluble UA that activates the NLRP3 inflammasome and pro-IL-1β transcription in macrophage-like J774.1 cells.
Collapse
Affiliation(s)
- Tomomi Notsu
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University, Yonago, Japan
| | - Yasutaka Kurata
- Department of Physiology II, Kanazawa Medical University, Uchinada, 920-0293, Japan.
| | - Haruaki Ninomiya
- Department of Biological Regulation, Tottori University Faculty of Medicine, Yonago, Japan
| | - Fikri Taufiq
- Department of Cardiology, Faculty of Medicine, Brawijaya University, Kota Malang, Jawa Timur, Indonesia
| | - Koji Komatsu
- Department of Psychiatry disease, Tottori University, Yonago, Japan
| | - Junichiro Miake
- Division of Pharmacology, Department of Pathophysiological and Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tatsuya Sawano
- Division of Pharmacology, Department of Pathophysiological and Therapeutic Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Motokazu Tsuneto
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University, Yonago, Japan
| | - Yasuaki Shirayoshi
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University, Yonago, Japan
| | - Ichiro Hisatome
- Department of Cardiology, National Hospital Organization Yonago Medical Center, Yonago, Japan
| |
Collapse
|
11
|
Tanaka Y, Nagoshi T, Takahashi H, Oi Y, Yasutake R, Yoshii A, Kimura H, Kashiwagi Y, Tanaka TD, Shimoda M, Yoshimura M. URAT1 is expressed in cardiomyocytes and dotinurad attenuates the development of diet-induced metabolic heart disease. iScience 2023; 26:107730. [PMID: 37694143 PMCID: PMC10483053 DOI: 10.1016/j.isci.2023.107730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/17/2023] [Accepted: 08/23/2023] [Indexed: 09/12/2023] Open
Abstract
We recently reported that the selective inhibition of urate transporter-1 (URAT1), which is primarily expressed in the kidneys, ameliorates insulin resistance by attenuating hepatic steatosis and improving brown adipose tissue function in diet-induced obesity. In this study, we evaluated the effects of dotinurad, a URAT1-selective inhibitor, on the hearts of high-fat diet (HFD)-fed obese mice for 16-20 weeks and on neonatal rat cardiomyocytes (NRCMs) exposed to palmitic acid. Outside the kidneys, URAT1 was also expressed in cardiomyocytes and indeed worked as a uric acid transporter. Dotinurad substantially attenuated HFD-induced cardiac fibrosis, inflammatory responses, and cardiac dysfunction. Intriguingly, among various factors related to the pathophysiology of diet-induced obesity, palmitic acid significantly increased URAT1 expression in NRCMs and subsequently induced apoptosis, oxidative stress, and inflammatory responses via MAPK pathway, all of which were reduced by dotinurad. These results indicate that URAT1 is a potential therapeutic target for metabolic heart disease.
Collapse
Affiliation(s)
- Yoshiro Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hirotake Takahashi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yuhei Oi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Rei Yasutake
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Akira Yoshii
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yusuke Kashiwagi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Toshikazu D. Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
12
|
Huang J, Dai M, He M, Bu W, Cao L, Jing J, Cao R, Zhang H, Men K. Treatment of Ulcerative Colitis by Cationic Liposome Delivered NLRP3 siRNA. Int J Nanomedicine 2023; 18:4647-4662. [PMID: 37605735 PMCID: PMC10440093 DOI: 10.2147/ijn.s413149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023] Open
Abstract
Purpose The abnormal activation of NLRP3 inflammasome is related to the occurrence and development of ulcerative colitis (UC). However, the ideal drug and delivery system remain important factors limiting the targeting of NLRP3 inflammasome in UC therapy. Gene therapy by delivering siRNA is effective in treating various diseases. Therefore, delivering siNLRP3 using an ideal vector for UC treatment is necessary. Materials and Methods Nanoparticles delivering siNLRP3 were developed based on cationic liposome (CLP/siNLRP3). Their ability to inhibit NLRP3 inflammasome activation was monitored using Western blot (WB) and Enzyme-linked Immunosorbent Assay (ELISA). The ASC oligomerization in LPS-primed peritoneal macrophages (PMs) was detected by WB and immunofluorescence. Moreover, we assessed the role of CLP/siNLRP3 on dextran sodium sulfate (DSS)-induced UC by examining NLRP3 levels, pro-inflammatory cytokines expression, and disease-associated index (DAI). Flow cytometry (FCM) was used to detect the contents of macrophages and T cells. Finally, we assessed the safety of CLP/siNLRP3. Results The prepared CLP was spherical, with a small particle size (94 nm) and low permeability. The CLP could efficiently protect siNLRP3 from degradation and then deliver siNLRP3 into PMs, inhibiting NLRP3 inflammasome activation. Also, the CLP/siNLRP3 could inhibit the secretion of mature IL-1β and IL-18 from PMs, thereby achieving a favorable anti-inflammation effect. In vivo, CLP/siNLRP3 could effectively alleviate intestinal injury in UC mice, which was attributed to down-regulating levels of IL-1β and IL-18, inhibiting infiltration of macrophages and other immune cells, and the polarization of M1 macrophages. Finally, pathological testing of tissue sections and blood biochemical tests showed no significant toxic effects of CLP/siNLRP3. Conclusion We introduced a prospective approach for the efficient delivery of siRNA in vitro and in vivo with high safety and stability, which was found to have great potential in treating NLRP3-driven diseases in an RNA-silencing manner.
Collapse
Affiliation(s)
- Jing Huang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan Province, 475004, People’s Republic of China
| | - Mengmeng Dai
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan Province, 475004, People’s Republic of China
| | - Mingxia He
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan Province, 475004, People’s Republic of China
| | - Weicheng Bu
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan Province, 475004, People’s Republic of China
| | - Liwen Cao
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan Province, 475004, People’s Republic of China
| | - Jing Jing
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan Province, 475004, People’s Republic of China
| | - Run Cao
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan Province, 475004, People’s Republic of China
| | - Hailong Zhang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan Province, 475004, People’s Republic of China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610044, People’s Republic of China
| |
Collapse
|
13
|
Chen X, You J, Zhou M, Ma H, Huang C. The association between serum uric acid and creatine phosphokinase in the general population: NHANES 2015-2018. BMC Cardiovasc Disord 2023; 23:296. [PMID: 37303058 DOI: 10.1186/s12872-023-03333-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 06/05/2023] [Indexed: 06/13/2023] Open
Abstract
BACKGROUND The association between serum creatine phosphokinase (CPK), a standard biochemical measure of acute myocardial infarction, and serum uric acid (sUA) has not been studied. This study aimed to determine the association between sUA and CPK in the general population of the US. METHODS Data from the National Health and Nutrition Examination Survey (NHANES) 2015-2018 were used, including a total of 8,431 subjects aged ≥ 30 years. Weighted multiple regression analysis was used to estimate the independent relationship between sUA and CPK. Fitted smoothing curves and weighted generalized additive models were also performed. RESULTS We found a positive relationship between sUA and CPK after adjusting for potential confounders. In subgroup analyses stratified by sex and race/ethnicity, sUA was positively correlated with CPK in each subgroup. The association between sUA and CPK followed an inverted U-shaped curve in females (turning point: sUA = 428.3 μmol/L). CONCLUSIONS Our study suggested that sUA level was positively correlated with CPK in the general population of the US. However, CPK increased with sUA until the turning point (sUA = 428.3 μmol/L) in females. Fundamental research and large sample prospective studies are needed to determine the exact mechanism of the association between sUA and CPK.
Collapse
Affiliation(s)
- Xinxin Chen
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jiuhong You
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Mei Zhou
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Hui Ma
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Huang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Pesent Address: West China Hospital, Sichuan University, No. 37, Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, China.
| |
Collapse
|
14
|
Wang Q, Peng D, Huang B, Men L, Jiang T, Huo S, Wang M, Guo J, Lv J, Lin L. Notopterol Ameliorates Hyperuricemia-Induced Cardiac Dysfunction in Mice. Pharmaceuticals (Basel) 2023; 16:ph16030361. [PMID: 36986461 PMCID: PMC10052463 DOI: 10.3390/ph16030361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/02/2023] Open
Abstract
Notopterol is a naturally occurring furanocoumarin compound found in the root of Notopterygium incisum. Hyperuricemia involves the activation of chronic inflammation and leads to cardiac damage. Whether notopterol has cardioprotective potential in hyperuricemia mice remains elusive. The hyperuricemic mouse model was constructed by administration of potassium oxonate and adenine every other day for six weeks. Notopterol (20 mg/kg) and allopurinol (10 mg/kg) were given daily as treatment, respectively. The results showed that hyperuricemia dampened heart function and reduced exercise capacity. Notopterol treatment improved exercise capacity and alleviated cardiac dysfunction in hyperuricemic mice. P2X7R and pyroptosis signals were activated both in hyperuricemic mice and in uric acid-stimulated H9c2 cells. Additionally, it was verified that inhibition of P2X7R alleviated pyroptosis and inflammatory signals in uric acid-treated H9c2 cells. Notopterol administration significantly suppressed expression levels of pyroptosis associated proteins and P2X7R in vivo and in vitro. P2X7R overexpression abolished the inhibition effect of notopterol on pyroptosis. Collectively, our findings suggested that P2X7R played a critical role in uric acid-induced NLRP3 inflammatory signals. Notopterol inhibited pyroptosis via inhibiting the P2X7R/NLRP3 signaling pathway under uric acid stimulation. Notopterol might represent a potential therapeutic strategy against pyroptosis and improve cardiac function in hyperuricemic mice.
Collapse
Affiliation(s)
- Qian Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dewei Peng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bingyu Huang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lintong Men
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tao Jiang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shengqi Huo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Moran Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Junyi Guo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiagao Lv
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Correspondence: or
| |
Collapse
|
15
|
Yang Y, Lin C, Zheng Q, Zhang L, Li Y, Huang Q, Wu T, Zhao Z, Li L, Luo J, Jiang Y, Zhang Q, Wang X, Xia C, Pang J. L-carnitine attenuated hyperuricemia-associated left ventricular remodeling through ameliorating cardiomyocytic lipid deposition. Front Pharmacol 2023; 14:1016633. [PMID: 36817129 PMCID: PMC9929955 DOI: 10.3389/fphar.2023.1016633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/06/2023] [Indexed: 02/04/2023] Open
Abstract
Hyperuricemia (HUA) is associated with left ventricular remodeling (LVR) and thereby causes the initiation and development of a large number of cardiovascular diseases. LVR is typically accompanied by cardiomyocyte energy metabolic disorder. The energy supply of cardiomyocytes is provided by glucose and fatty acid (FA) metabolism. Currently, the effect of HUA on cardiomyocytic FA metabolism is unclear. In this study, we demonstrate that UA-induced cardiomyocyte injury is associated with cytoplasmic lipid deposition, which can be ameliorated by the FA metabolism-promoting drug L-carnitine (LC). UA suppresses carnitine palmitoyl transferase 1B (CPT1B), thereby inhibiting FA transport into the mitochondrial inner matrix for elimination. LC intervention can ameliorate HUA-associated left ventricular anterior wall thickening in mice. This study showed that FA transport dysfunction plays is a critical mechanism in both cardiomyocytic injury and HUA-associated LVR and promoting cytoplasmic FA transportation through pharmacological treatment by LC is a valid strategy to attenuate HUA-associated LVR.
Collapse
Affiliation(s)
- Yang Yang
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong, China,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China,NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Cuiting Lin
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiang Zheng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Leqi Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongmei Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinghua Huang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zean Zhao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Lu Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Luo
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanqing Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Qun Zhang
- Good Clinical Practice Development, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xing Wang
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong, China,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Chenglai Xia
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong, China,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China,*Correspondence: Jianxin Pang, ; Chenglai Xia,
| | - Jianxin Pang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China,*Correspondence: Jianxin Pang, ; Chenglai Xia,
| |
Collapse
|
16
|
Rodriguez-Iturbe B, Johnson RJ, Lanaspa MA, Nakagawa T, Garcia-Arroyo FE, Sánchez-Lozada LG. Sirtuin deficiency and the adverse effects of fructose and uric acid synthesis. Am J Physiol Regul Integr Comp Physiol 2022; 322:R347-R359. [PMID: 35271385 PMCID: PMC8993531 DOI: 10.1152/ajpregu.00238.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 12/17/2022]
Abstract
Fructose metabolism and hyperuricemia have been shown to drive insulin resistance, metabolic syndrome, hepatic steatosis, hypertension, inflammation, and innate immune reactivity in experimental studies. We suggest that these adverse effects are at least in part the result of suppressed activity of sirtuins, particularly Sirtuin1. Deficiency of sirtuin deacetylations is a consequence of reduced bioavailability of its cofactor nicotinamide adenine dinucleotide (NAD+). Uric acid-induced inflammation and oxidative stress consume NAD+ and activation of the polyol pathway of fructose and uric acid synthesis also reduces the NAD+-to-NADH ratio. Variability in the compensatory regeneration of NAD+ could result in variable recovery of sirtuin activity that may explain the inconsistent benefits of treatments directed to reduce uric acid in clinical trials. Here, we review the pathogenesis of the metabolic dysregulation driven by hyperuricemia and their potential relationship with sirtuin deficiency. In addition, we discuss therapeutic options directed to increase NAD+ and sirtuins activity that may improve the adverse effects resulting from fructose and uric acid synthesis.
Collapse
Affiliation(s)
- Bernardo Rodriguez-Iturbe
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán," Mexico City, Mexico
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Denver, Colorado
- Kidney Disease Division, Rocky Mountain Regional Veterans Affairs Medical Center, Denver, Colorado
| | - Miguel A Lanaspa
- Division of Nephrology and Hypertension, Oregon Health and Science University, Portland, Oregon
| | | | - Fernando E Garcia-Arroyo
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| | - Laura G Sánchez-Lozada
- Departments of Cardio-Renal Physiopathology Instituto Nacional de Cardiología "Ignacio Chavez," Mexico City, Mexico
| |
Collapse
|
17
|
Ren W, Wu J, Wu Z, Yang S, Jiang X, Xu M, Wu B, Xie C, He J, Yu X. Serum Uric Acid Levels at Admission Could Predict the Chronic Post-stroke Fatigue. Front Nutr 2022; 9:850355. [PMID: 35273991 PMCID: PMC8902642 DOI: 10.3389/fnut.2022.850355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background Post-stroke fatigue (PSF) is a frequent complication of stroke. Serum uric acid (SUA) is frequently thought to be a risk factor for stroke. This study aimed to investigate whether SUA also played a role in PSF. Methods Subjects with ischemic stroke were screened from The First Affiliated Hospital of Wenzhou Medical University between January 2020 and October 2020. Patients' fatigue symptoms were assessed by the Fatigue severity scale (FSS). To investigate the relationship between SUA and PSF, binary logistic regression analysis was conducted, with the confounders being controlled. SUA levels were divided into four layers (Q1 ≤ 245 μmol/L; Q2 246–308 μmol/L; Q3 309–365 μmol/L; Q4 ≥366 μmol/L) based on the quartiles. Results SUA levels were significantly higher in the PSF group (345.96 ± 73.78 μmol/L) than the non-PSF group (295.97 ± 87.8 μmol/L, P < 0.001). There were no differences in any other variables between these two groups. After adjusting the confounders, the risk of PSF in the Q4 layer (≥366 μmol/L) was 6.05 times (95% CI 1.79–20.43, P = 0.004) higher than that in Q1 (≤245 μmol/L). Conclusion High SUA at admission was an independent risk factor for fatigue 1 year after stroke onset. High SUA (≥366 μmol/L) during stroke deserves more attention, and active control of high SUA levels may be beneficial to reduce the incidence of PSF in the chronic stage following stroke.
Collapse
Affiliation(s)
- Wenwei Ren
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Junxin Wu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Zijing Wu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Shuang Yang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xiaofang Jiang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minjie Xu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Beilan Wu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Caixia Xie
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Jincai He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Yu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,Peking University Institute of Mental Health (Sixth Hospital), Beijing, China.,National Clinical Research Center for Mental Disorders and Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China.,Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, China
| |
Collapse
|
18
|
Sun ZR, Liu HR, Hu D, Fan MS, Wang MY, An MF, Zhao YL, Xiang ZM, Sheng J. Ellagic Acid Exerts Beneficial Effects on Hyperuricemia by Inhibiting Xanthine Oxidase and NLRP3 Inflammasome Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:12741-12752. [PMID: 34672194 DOI: 10.1021/acs.jafc.1c05239] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Hyperuricemia is a metabolic disease caused by impaired uric acid (UA) metabolism. Ellagic acid (EA) is a natural small-molecule polyphenolic compound with known antioxidative and anti-inflammatory properties. Here, we evaluated the regulatory effects of EA on hyperuricemia and explored the underlying mechanisms. We found that EA is an effective xanthine oxidase (XOD) inhibitor (IC50 = 165.6 μmol/L) and superoxide anion scavenger (IC50 = 27.66 μmol/L). EA (5 and 10 μmol/L) treatment significantly and dose-dependently reduced UA levels in L-O2 cells; meanwhile, intraperitoneal EA administration (50 and 100 mg/kg) also significantly reduced serum XOD activity and UA levels in hyperuricemic mice and markedly improved their liver and kidney histopathology. EA treatment significantly reduced the degree of foot edema and inhibited the expression of NLPR3 pathway-related proteins in foot tissue of monosodium urate (MSU)-treated mice. The anti-inflammatory effect was also observed in lipopolysaccharide-stimulated RAW-264.7 cells. Furthermore, EA significantly inhibited the expressions of XOD and NLRP3 pathway-related proteins (TLR4, p-p65, caspase-1, TNF-α, and IL-18) in vitro and in vivo. Our results indicated that EA exerts ameliorative effects in experimental hyperuricemia and foot edema via regulating the NLRP3 signaling pathway and represents a promising therapeutic option for the management of hyperuricemia.
Collapse
Affiliation(s)
- Ze-Rui Sun
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650224, P. R. China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650224, P. R. China
| | - Hua-Rong Liu
- College of Health Nursing Sciences, Yunnan Open University, Kunming 650223, P. R. China
| | - Di Hu
- Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, P. R. China
| | - Mao-Si Fan
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650224, P. R. China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650224, P. R. China
| | - Ming-Yue Wang
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650224, P. R. China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650224, P. R. China
| | - Meng-Fei An
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650224, P. R. China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650224, P. R. China
| | - Yun-Li Zhao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education; Yunnan Provincial Center for Research & Development of Natural Products; School of Chemical Science and Technology, Yunnan University, Kunming 650091, P. R. China
| | - Ze-Min Xiang
- College of Science, Yunnan Agricultural University, Kunming 650224, P. R. China
| | - Jun Sheng
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650224, P. R. China
- College of Science, Yunnan Agricultural University, Kunming 650224, P. R. China
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming 650224, P. R. China
| |
Collapse
|
19
|
Li D, Wang L, Ou J, Wang C, Zhou J, Lu L, Wu Y, Gao J. Reactive oxygen species induced by uric acid promote NRK‑52E cell apoptosis through the NEK7‑NLRP3 signaling pathway. Mol Med Rep 2021; 24:729. [PMID: 34414459 PMCID: PMC8383041 DOI: 10.3892/mmr.2021.12368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/29/2021] [Indexed: 01/07/2023] Open
Abstract
Increasing uric acid (UA) could induce renal tubular epithelial cell (NRK-52E) injury. However, the specific mechanism by which UA induces renal tubular epithelial cell injury remains unknown. It was hypothesized that UA induces renal tubular epithelial cell injury through reactive oxygen species (ROS) and the Never in mitosis gene A (NIMA)-related kinase 7 (NEK7)/NLR family pyrin domain containing 3 (NLRP3) signaling pathway. TUNEL assay and flow cytometry were applied to measure apoptosis, and the results of the present study showed that UA treatment induced apoptosis of NRK-52E cells in a concentration-dependent manner. Western blotting was performed to determine the expression levels of cleaved caspase-3, Bax and Bcl-xl, it was found that levels were significantly increased after UA treatment in NRK-52E cells. ROS and apoptosis were predominantly induced in NRK-52E cells and there was an association between ROS and apoptosis. Enhanced expression of NEK7, NLRP3, apoptosis-associated speck-like and caspase-1 were observed in NRK-52E cells treated with UA. The ROS inhibitor, N-acetyl-l-cysteine, exerted a protective effect on the UA-induced apoptosis of tubular epithelial cells by reducing excess ROS production, which significantly inhibited NEK7 and NLRP3 inflammasome activation. These results indicated that UA activates ROS and induces apoptosis of NRK-52E cells. The mechanism might be related to the regulation of the NEK7/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Dongdong Li
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, P.R. China
| | - Luobing Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, P.R. China
| | - Jiaoying Ou
- Department of Internal Medicine, Shanghai TCM‑Integrated Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200082, P.R. China
| | - Chuanxu Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, P.R. China
| | - Jiabao Zhou
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, P.R. China
| | - Lili Lu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, P.R. China
| | - Yanshneg Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, P.R. China
| | - Jiandong Gao
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
20
|
Timosaponin alleviates oxidative stress in rats with high fat diet-induced obesity via activating Nrf2/HO-1 and inhibiting the NF-κB pathway. Eur J Pharmacol 2021; 909:174377. [PMID: 34302815 DOI: 10.1016/j.ejphar.2021.174377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/11/2021] [Accepted: 07/21/2021] [Indexed: 12/29/2022]
Abstract
Anemarrhena asphodeloides originated from the rhizome of Liliaceae Anemarrhena asphodeloides. One of the active pharmacological components of Anemarrhena asphodeloides is timosaponin (TSA), which reduces blood lipids and shows antioxidation and anti-inflammatory effects, but its mechanism is unclear. The objective of this study was to investigate the effect of TSA on oxidative stress induced by a long-term high-fat diet in obese rats. Body weight and the obesity index of the rats were measured during the experiment. Total antioxidant capacity (T-AOC), malondialdehyde (MDA), and glutathione peroxidase (GSH-Px) were used to detect oxidative stress indexes in serum and liver tissue. To observe the effect of TSA on the liver and adipose tissue of rats with oxidative stress, hematoxylin & eosin (H&E) staining was used. The p-NF-κB, NAD(P)H: quinone oxidoreductase 1 (NQO-1), Heme oxygenase 1 (HO-1), and Nrf2 in Nrf2/HO-1 and NF-κB pathways were assayed by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot. TSA was found to improve oxidative stress in obese rats by reducing MDA levels and increasing T-AOC and GSH-Px levels. Histological examination revealed that TSA effectively attenuated liver damage and improved obesity in rats. TSA was found to down-regulate the protein level of p-NF-κB and up-regulate the protein level of Nrf2/HO-1. These results suggested that TSA could effectively block inflammation and dyslipidemia in obese rats, thus improving oxidative stress, and its mechanism could be related to the Nrf2/HO-1 and NF-κB pathways.
Collapse
|
21
|
Li L, Li Y, Luo J, Jiang Y, Zhao Z, Chen Y, Huang Q, Zhang L, Wu T, Pang J. Resveratrol, a novel inhibitor of GLUT9, ameliorates liver and kidney injuries in a D-galactose-induced ageing mouse model via the regulation of uric acid metabolism. Food Funct 2021; 12:8274-8287. [PMID: 34180933 DOI: 10.1039/d1fo00538c] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Accumulating evidence has shown that chronic injection of d-galactose (d-gal) can mimic natural ageing and induce liver and kidney injury. Previous studies showed that d-gal increased uric acid (UA) levels in mice. The increase in UA levels caused inflammation, accelerated oxidative stress, and aggravated liver and kidney injury. Oxidative stress and inflammation play vital roles in the ageing process. Therefore, reducing the levels of UA in ageing mice improved liver and kidney injury. Glucose transporter 9 (GLUT9) is responsible for the reabsorption of UA in the body, and its inhibition helps downregulate UA levels. The present study investigated the UA-lowering activity of the GLUT9 inhibitor resveratrol (RSV) using the patch clamping technique established in our laboratory in vitro. This research is the first study to demonstrate that RSV effectively inhibits UA uptake via GLUT9 (IC50 = 68.77 μM) in vitro. An in vivo study was also performed to investigate the possible protective effect of RSV on d-gal-induced liver and kidney injury. RSV significantly reduced serum UA levels via the downregulation of GLUT9 mRNA and protein expression and promoted the excretion of excess UA through urine. Biochemical analysis showed that RSV significantly downregulated abnormal increases in serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN) and creatinine (CRE) caused by long-term d-gal treatment, which effectively improved pathological damage, increased superoxide dismutase (SOD) activity and decreased the content of malondialdehyde (MDA) in the liver and kidneys. RSV also downregulated the expression of the inflammatory cytokines, interleukin IL-6, IL-1β and tumor necrosis factor (TNF)-α in the liver and kidneys of ageing mice. Our findings provide new insights into the treatment strategies for ageing-induced liver and kidney injury and reveal a new mechanism of RSV-induced reduction in UA levels in ageing individuals.
Collapse
Affiliation(s)
- Lu Li
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yongmei Li
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Jian Luo
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yanqing Jiang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Zean Zhao
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yanyu Chen
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Qinghua Huang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Leqi Zhang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Ting Wu
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
22
|
Zhang H, Ma Y, Cao R, Wang G, Li S, Cao Y, Zhang H, Liu M, Liu G, Zhang J, Li S, Wang Y, Ma Y. Soluble uric acid induces myocardial damage through activating the NLRP3 inflammasome. J Cell Mol Med 2020; 24:8849-8861. [PMID: 32558367 PMCID: PMC7412683 DOI: 10.1111/jcmm.15523] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 04/25/2020] [Accepted: 05/24/2020] [Indexed: 01/29/2023] Open
Abstract
Uric acid crystal is known to activate the NLRP3 inflammasome and to cause tissue damages, which can result in many diseases, such as gout, chronic renal injury and myocardial damage. Meanwhile, soluble uric acid (sUA), before forming crystals, is also related to these diseases. This study was carried out to investigate whether sUA could also activate NLRP3 inflammasome in cardiomyocytes and to analyse the mechanisms. The cardiomyocyte activity was monitored, along with the levels of mature IL-1β and caspase-1 from H9c2 cells following sUA stimulus. We found that sUA was able to activate NLRP3 inflammasome, which was responsible for H9c2 cell apoptosis induced by sUA. By elevating TLR6 levels and then activating NF-κB/p65 signal pathway, sUA promoted NLRP3, pro-caspase 1 and pro-IL-1β production and provided the first signal of NLRP3 inflammasome activation. Meanwhile, ROS production regulated by UCP2 levels also contributed to NLRP3 inflammasome assembly and subsequent caspase 1 activation and mature IL-1β secretion. In addition, the tlr6 knockdown rats suffering from hyperuricemia showed the lower level of IL-1β and an ameliorative cardiac function. These findings suggest that sUA activates NLRP3 inflammasome in cardiomyocytes and they may provide one therapeutic strategy for myocardial damage induced by sUA.
Collapse
Affiliation(s)
- Hailong Zhang
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Yuting Ma
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Run Cao
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Guanli Wang
- Clinical LaboratoryHuaihe HospitalHenan UniversityKaifengChina
| | - Shaowei Li
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Yue Cao
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Hao Zhang
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Meichen Liu
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Guangchao Liu
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Jun Zhang
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Shulian Li
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Yaohui Wang
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| | - Yuanfang Ma
- Joint National Laboratory for Antibody Drug EngineeringKey Laboratory of Cellular and Molecular Immunology of Henan ProvinceSchool of Basic MedicineHenan UniversityKaifengChina
| |
Collapse
|