1
|
Huang S, Liang H, Chen Y, Liu C, Luo P, Wang H, Du Q. Hypoxanthine ameliorates diet-induced insulin resistance by improving hepatic lipid metabolism and gluconeogenesis via AMPK/mTOR/PPARα pathway. Life Sci 2024; 357:123096. [PMID: 39369847 DOI: 10.1016/j.lfs.2024.123096] [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: 06/11/2024] [Revised: 09/09/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
AIM Insulin resistance (IR) is a pivotal metabolic disorder associated with type 2 diabetes and metabolic syndrome. This study investigated the potential of hypoxanthine (Hx), a purine metabolite and uric acid precursor, in ameliorating IR and regulating hepatic glucose and lipid metabolism. METHODS We utilized both in vitro IR-HepG2 cells and in vivo diet-induced IR mice to investigate the impact of Hx. The HepG2 cells were treated with Hx to evaluate its effects on glucose production and lipid deposition. Activity-based protein profiling (ABPP) was applied to identify Hx-target proteins and the underlying pathways. In vivo studies involved administration of Hx to IR mice, followed by assessments of IR-associated indices, with explores on the potential regulating mechanisms on hepatic glucose and lipid metabolism. KEY FINDINGS Hx intervention significantly reduced glucose production and lipid deposition in a dose-dependent manner without affecting cell viability in IR-HepG2 cells. ABPP identified key Hx-target proteins engaged in fatty acid and pyruvate metabolism. In vivo, Hx treatment reduced IR severities, as evidenced by decreased HOMA-IR, fasting blood glucose, and serum lipid profiles. Histological assessments confirmed reduced liver lipid deposition. Mechanistic insights revealed that Hx suppresses hepatic gluconeogenesis and fatty acid synthesis, and promotes fatty acid oxidation via the AMPK/mTOR/PPARα pathway. SIGNIFICANCE This study delineates a novel role of Hx in regulating hepatic metabolism, offering a potential therapeutic strategy for IR and associated metabolic disorders. The findings provide a foundation for further investigation into the role of purine metabolites in metabolic regulation and their clinical implications.
Collapse
Affiliation(s)
- Sizhe Huang
- Centre of General Practice, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China
| | - Hengmiao Liang
- Centre of General Practice, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China
| | - Yuting Chen
- Department of Laboratory Medicine, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Ce Liu
- Department of Laboratory Medicine, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China
| | - Piao Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| | - Huijun Wang
- Centre of General Practice, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China; Department of Laboratory Medicine, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China.
| | - Qingfeng Du
- Centre of General Practice, the Seventh Affiliated Hospital, Southern Medical University, Foshan 528000, Guangdong, PR China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
2
|
Liu C, Ruan F, Chen Z, Han J, Ding X, Han C, Ye L, Yang C, Yu Y, Zuo Z, He C. Phenanthrene-induced hyperuricemia with intestinal barrier damage and the protective role of theabrownin: Modulation by gut microbiota-mediated bile acid metabolism. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:174923. [PMID: 39047823 DOI: 10.1016/j.scitotenv.2024.174923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/24/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Hyperuricemia is prevalent globally and potentially linked to environmental pollution. As a typical persistent organic pollutant, phenanthrene (Phe) poses threats to human health through biomagnification. Although studies have reported Phe-induced toxicities to multiple organs, its impact on uric acid (UA) metabolism remains unclear. In this study, data mining on NHANES 2001-2016 indicated a positive correlation between Phe exposure and the occurrence of hyperuricemia in population. Subsequently, adolescent Balb/c male mice were orally exposed to Phe at a dosage of 10 mg/kg bw every second day for 7 weeks, resulting in dysfunction of intestinal UA excretion and disruption of the intestinal barrier. Utilizing intestinal organoids, 16S rRNA sequencing of gut microbiota, and targeted metabolomic analysis, we further revealed that an imbalance in bile acid metabolism derived from gut microbiota might mediate the intestinal barrier damage. Additionally, the tea extract theabrownin (TB) effectively improved Phe-induced hyperuricemia and intestinal dysfunction at a dose of 320 mg/kg bw per day. In conclusion, this study demonstrates that Phe exposure is positively associated with hyperuricemia and intestinal damage, which provides new insights into the toxic effects induced by Phe. Furthermore, the present study proposes that supplementation with TB would be a healthy and effective improvement strategy for patients with hyperuricemia and intestinal injury caused by environmental factors.
Collapse
Affiliation(s)
- Changqian Liu
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Fengkai Ruan
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhiyuan Chen
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jianrong Han
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaoyan Ding
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Changshun Han
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Lingxiao Ye
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Chunyan Yang
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yi Yu
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
3
|
Liu M, Shen J, Chen X, Dawuti T, Xiao H. Evaluating renal injury characteristics in different rat models of hyperuricemia and elucidating pathological molecular mechanisms via serum metabolomics. Front Pharmacol 2024; 15:1433991. [PMID: 39286632 PMCID: PMC11403331 DOI: 10.3389/fphar.2024.1433991] [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: 05/16/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Hyperuricemia has emerged as a significant global health concern, closely associated with various metabolic disorders. The adverse effects frequently observed with current pharmacological treatments for hyperuricemia highlight the urgent need for reliable animal models to elucidate the disease's pathophysiological mechanisms, thereby facilitating the development of safer and more effective therapies. In this study, we established three rat models of hyperuricemia using potassium oxonate, either alone or in combination with fructose and adenine. Each model exhibited distinct pathological changes, with the combination of potassium oxonate, fructose, and adenine causing significantly more severe damage to liver and kidney functions than potassium oxonate alone. Serum metabolomics analyses revealed profound dysregulation in the metabolic pathways of purine, pyrimidines, and glutathione, underscoring the pivotal role of oxidative stress in the progression of hyperuricemia. We identified key biomarkers such as orotidine, ureidosuccinic acid, uracil, and pseudouridine, which are associated with uric acid-induced damage to hepatic and renal systems. MetOrigin tracing analysis further revealed that differential metabolites related to hyperuricemia are primarily involved in host-microbiome co-metabolic pathways, particularly in purine metabolism, with bacterial phyla such as Pseudomonadota, Actinomycetota, and Ascomycota being closely linked to the critical metabolic processes of uric acid production. These findings not only enhance our understanding of the pathogenic mechanisms underlying hyperuricemia but also provide a robust experimental model foundation for the development of innovative treatment strategies.
Collapse
Affiliation(s)
- Mengwen Liu
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Jing Shen
- School of Public Health, Xinjiang Medical University, Urumqi, China
- Key Laboratory of Environmental Exposome, Xinjiang Medical University, Urumqi, China
| | - Xuanshi Chen
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | | | - Hui Xiao
- School of Public Health, Xinjiang Medical University, Urumqi, China
- Key Laboratory of Environmental Exposome, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
4
|
Mehmood A, Iftikhar A, Chen X. Food-derived bioactive peptides with anti-hyperuricemic activity: A comprehensive review. Food Chem 2024; 451:139444. [PMID: 38678657 DOI: 10.1016/j.foodchem.2024.139444] [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: 10/06/2023] [Revised: 04/01/2024] [Accepted: 04/20/2024] [Indexed: 05/01/2024]
Abstract
Hyperuricemia (HU) is a metabolic disorder caused by the overproduction or underexcretion of uric acid (UA) in the human body. Several approved drugs for the treatment of HU are available in the market; however, all these allopathic drugs exhibit multiple side effects. Therefore, the development of safe and effective anti-HU drugs is an urgent need. Natural compounds derived from foods and plants have the potential to decrease UA levels. Recently, food-derived bioactive peptides (FBPs) have gained attention as a functional ingredient owing to their biological activities. In the current review, we aim to explore the urate-lowering potential and the underlying mechanisms of FBPs. We found that FBPs mitigate HU by reducing blood UA levels through inhibiting key enzymes such as xanthine oxidase, increasing renal UA excretion, inhibiting renal UA reabsorption, increasing anti-oxidant activities, regulating inflammatory mediators, and addressing gut microbiota dysbiosis. In conclusion, FBPs exhibit strong potential to ameliorate HU.
Collapse
Affiliation(s)
- Arshad Mehmood
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, PR China; Institute of Food Physical Processing, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, PR China
| | - Asra Iftikhar
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, The University of Faisalabad, Faisalabad 38000, Pakistan and Akhtar Saeed College of Pharmacy, Rawalpindi, Pakistan
| | - Xiumin Chen
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, PR China; Institute of Food Physical Processing, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, PR China; International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
5
|
Jiang T, Weng Q, Zhang Y, Zhang W, Doherty M, Sarmanova A, Yang Z, Yang T, Li J, Liu K, Wang Y, Obotiba AD, Zeng C, Lei G, Wei J. Association Between Hyperuricemia and Ultrasound-Detected Hand Synovitis. Arthritis Care Res (Hoboken) 2024; 76:1187-1194. [PMID: 38570913 DOI: 10.1002/acr.25342] [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: 08/06/2023] [Revised: 02/15/2024] [Accepted: 04/02/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVE Although hand synovitis is prevalent in the older population, the etiology remains unclear. Hyperuricemia, a modifiable metabolic disorder, may serve as an underlying mechanism of hand synovitis, but little is known about their relationship. We assessed the association between hyperuricemia and hand synovitis in a large population-based sample. METHODS We performed a cross-sectional study in Longshan County, Hunan Province, China. Hyperuricemia was defined as a serum urate level >420 μmol/L in men and >360 μmol/L in women. Ultrasound examinations were performed on both hands of 4,080 participants, and both gray-scale synovitis and the Power Doppler signal (PDS) were assessed using semiquantitative scores (grades 0-3). We evaluated the association of hyperuricemia with hand gray-scale synovitis (grade ≥2) and PDS (grade ≥1), respectively, adjusting for age, sex, and body mass index. RESULTS All required assessments for analysis were available for 3,286 participants. The prevalence of hand gray-scale synovitis was higher among participants with hyperuricemia (30.0%) than those with normouricemia (23.3%), with an adjusted odds ratio (aOR) of 1.28 (95% confidence interval [CI] 1.00-1.62). Participants with hyperuricemia also had a higher prevalence of PDS (aOR 2.36; 95% CI 1.15-4.81). Furthermore, hyperuricemia positively associated, both at the hand and joint levels, with the presence of gray-scale synovitis (aOR 1.27; 95% CI 1.00-1.60 and adjusted prevalence ratio [aPR] 1.26; 95% CI 1.10-1.44, respectively) and PDS (aOR 2.35; 95% CI 1.15-4.79 and aPR 2.34; 95% CI 1.28-4.30, respectively). CONCLUSION This population-based study provides more evidence for a positive association between hyperuricemia and prevalent hand synovitis.
Collapse
Affiliation(s)
- Ting Jiang
- Xiangya Hospital, Central South University, Changsha, China, the University of Nottingham, and Pain Centre Versus Arthritis UK, Nottingtham, United Kingdom
| | - Qianlin Weng
- Xiangya Hospital, Central South University, Changsha, China
| | - Yuqing Zhang
- Massachusetts General Hospital, Harvard Medical School, Boston
| | - Weiya Zhang
- University of Nottingham and Pain Centre Versus Arthritis UK, Nottingham, United Kingdom
| | - Michael Doherty
- University of Nottingham and Pain Centre Versus Arthritis UK, Nottingham, United Kingdom
| | | | - Zidan Yang
- Xiangya Hospital, Central South University, Changsha, China
| | - Tuo Yang
- Xiangya Hospital, Central South University, Changsha, China, the University of Nottingham, and Pain Centre Versus Arthritis UK, Nottingtham, United Kingdom
| | - Jiatian Li
- Xiangya Hospital, Central South University, Changsha, China
| | - Ke Liu
- Xiangya Hospital, Central South University, Changsha, China
| | - Yuqing Wang
- Xiangya Hospital, Central South University, Changsha, China
| | | | - Chao Zeng
- Xiangya Hospital, Central South University, Changsha, China
| | - Guanghua Lei
- Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wei
- Xiangya Hospital, Central South University, and Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
6
|
Zhang M, Hussain A, Hu B, Yang H, Li C, Guo S, Han X, Li B, Dai Y, Cao Y, Chi H, Weng Y, Qin CF, Huang Y. Atavistic strategy for the treatment of hyperuricemia via ionizable liposomal mRNA. Nat Commun 2024; 15:6463. [PMID: 39085241 PMCID: PMC11292028 DOI: 10.1038/s41467-024-50752-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Hyperuricemia is associated with an increased risk of gout, hypertension, diabetes, and cardiovascular diseases. Most mammals maintain normal serum uric acid (SUA) via urate oxidase (Uox), an enzyme that metabolizes poorly-soluble UA to highly-soluble allantoin. In contrast, Uox became a pseudogene in humans and apes over the long course of evolution. Here we demonstrate an atavistic strategy for treating hyperuricemia based on endogenous expression of Uox in hepatocytes mediated by mRNA (mUox) loaded with an ionizable lipid nanoparticle termed iLAND. mUox@iLAND allows effective transfection and protein expression in vitro. A single dose of mUox@iLAND lowers SUA levels for several weeks in two female murine models, including a novel long-lasting model, which is also confirmed by metabolomics analysis. Together with the excellent safety profiles observed in vivo, the proposed mRNA agent demonstrates substantial potential for hyperuricemia therapy and the prevention of associated conditions.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Bo Hu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Chunhui Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Shuai Guo
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Xiaofeng Han
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
- Rigerna Therapeutics Co. Ltd., Beijing, China
| | - Bei Li
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Yuhong Cao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, China
| | - Hang Chi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuhua Weng
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
7
|
Sun H, Wang X, Li Y, Shen Y, Zhang L, Xu Y, Liu J, Fan X. Targeting Liver Xor by GalNAc-siRNA Is an Effective Strategy for Hyperuricemia Therapy. Pharmaceutics 2024; 16:938. [PMID: 39065635 PMCID: PMC11279964 DOI: 10.3390/pharmaceutics16070938] [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: 04/09/2024] [Revised: 06/25/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Hyperuricemia, i.e., increased plasma uric acid concentration, is a common problem in clinical practice, leading to gout or nephrolithiasis, and is associated with other disorders, such as metabolic syndrome, cardiovascular disease, and chronic renal disease. Xanthine oxidoreductase (XOR) is a critical rate-limiting enzyme involved in uric acid synthesis and a promising target for hyperuricemia therapy. However, XOR inhibitors currently face clinical problems such as a short half-life and side effects. Here, we found that specifically targeting liver Xor with GalNAc-siRNAs had a good therapeutic effect on hyperuricemia. First, siRNAs were designed to target various sites in the homologous region between Homo sapiens and Mus musculus Xor mRNA and were screened in primary mouse hepatocytes. Then, the siRNAs were modified to increase their stability in vivo and conjugated with GalNAc for liver-specific delivery. The effects of GalNAc-siRNAs were evaluated in three hyperuricemia mouse models, including potassium oxonate and hypoxanthine administration in WT and humanized XDH mice and Uox knockout mice. Febuxostat, a specific XOR inhibitor used for hyperuricemia treatment, was used as a positive control. Targeting liver Xor with GalNAc-siRNAs by subcutaneous administration reduced plasma uric acid levels, uric acid accumulation in the kidney, renal inflammation, and fibrosis, thereby alleviating kidney damage in hyperuricemia mouse models without hepatoxicity. The results demonstrated that targeting liver Xor with GalNAc-siRNAs was a promising strategy for hyperuricemia therapy.
Collapse
Affiliation(s)
- Huiyan Sun
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinxia Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yongqiang Li
- Shanghai Synvida Biotechnology Co., Ltd., Shanghai 201203, China
| | - Yingzhi Shen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Synvida Biotechnology Co., Ltd., Shanghai 201203, China
| | - Xuemei Fan
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
8
|
Zeng L, Shali S, Gao Y, Du X, Zhu X, Li L, Dai Y, Zhou P. CRISPR/Cas9 Mediated Deletion of the Uox Gene Generates a Mouse Model of Hyperuricemia with Multiple Complications. J Cardiovasc Transl Res 2024:10.1007/s12265-024-10526-6. [PMID: 38856882 DOI: 10.1007/s12265-024-10526-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024]
Abstract
Hyperuricemia is a common metabolic disorder with severe complications. We aimed to develop a mouse model for spontaneous hyperuricemia. Uox-/- mouse model was generated on C57BL/6J background by deleting exon 2-4 of Uox using the CRISPR/Cas9 system. The prototypic Uox -/-mice had 5.5-fold increased serum uric acid (1351.04±276.58μmol/L) as compared to the wild type mice (P<0.0001), but died by 4 weeks. After allopurinol (3ug/g) intervention, they all survived > 8 weeks. The serum uric acid was 612.55±146.98μmol/L in the 8-week-old allopurinol-rescued Uox -/-mice, which manifested multiple complications including severe renal insufficiency, hypertension, left ventricular remodeling and systolic dysfunction, aortic endothelial dysfunction, hepatic steatosis and elevated liver enzymes, as well as hyperglycemia and hypercholesteremia. The present Uox-/- mice developed spontaneous hyperuricemia complicated with urate nephropathy, cardiovascular disease and cardiometabolic disorders, and may provide a novel tool to study hyperuricemia associated early-onset cardiovascular disorders in human.
Collapse
Affiliation(s)
- Linzi Zeng
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shalaimaiti Shali
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, China.
| | - Yabiao Gao
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xingchen Du
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Xiaoxia Zhu
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Lin Li
- Department of Nephrology, Shanghai Changzheng Hospital, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yuxiang Dai
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, China.
| | - Ping Zhou
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Wei W, Ma S, Fu B, Song R, Guo H. Human-specific insights into candidate genes and boosted discoveries of novel loci illuminate roles of neuroglia in reading disorders. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12899. [PMID: 38752599 PMCID: PMC11097622 DOI: 10.1111/gbb.12899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024]
Abstract
Reading disorders (RD) are human-specific neuropsychological conditions associated with decoding printed words and/or reading comprehension. So far only a handful of candidate genes segregated in families and 42 loci from genome-wide association study (GWAS) have been identified that jointly provided little clues of pathophysiology. Leveraging human-specific genomic information, we critically assessed the RD candidates for the first time and found substantial human-specific features within. The GWAS candidates (i.e., population signals) were distinct from the familial counterparts and were more likely pleiotropic in neuropsychiatric traits and to harbor human-specific regulatory elements (HSREs). Candidate genes associated with human cortical morphology indeed showed human-specific expression in adult brain cortices, particularly in neuroglia likely regulated by HSREs. Expression levels of candidate genes across human brain developmental stages showed a clear pattern of uplifted expression in early brain development crucial to RD development. Following the new insights and loci pleiotropic in cognitive traits, we identified four novel genes from the GWAS sub-significant associations (i.e., FOXO3, MAPT, KMT2E and HTT) and the Semaphorin gene family with functional priors (i.e., SEMA3A, SEMA3E and SEMA5B). These novel genes were related to neuronal plasticity and disorders, mostly conserved the pattern of uplifted expression in early brain development and had evident expression in cortical neuroglial cells. Our findings jointly illuminated the association of RD with neuroglia regulation-an emerging hotspot in studying neurodevelopmental disorders, and highlighted the need of improving RD phenotyping to avoid jeopardizing future genetic studies of RD.
Collapse
Affiliation(s)
- Wen‐Hua Wei
- Centre for Biostatistics, Division of Population Health, Health Services Research and Primary CareThe University of ManchesterManchesterUK
| | - Shaowei Ma
- Hebei Key Laboratory of Children's Cognition and Digital Education and School of Foreign LanguagesLangfang Normal UniversityLangfangChina
| | - Bo Fu
- School of Data ScienceFudan UniversityShanghaiChina
| | - Ranran Song
- Department of Maternal and Child Health and MOE (Ministry of Education) Key Laboratory of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hui Guo
- Centre for Biostatistics, Division of Population Health, Health Services Research and Primary CareThe University of ManchesterManchesterUK
| |
Collapse
|
10
|
Liu WW, Dong HJ, Zhang Z, Ma XH, Liu S, Huang W, Wang X. Analyzing chemical composition of Sargentodoxae caulis water extract and their hypouricemia effect in hyperuricemic mice. Fitoterapia 2024; 175:105926. [PMID: 38537887 DOI: 10.1016/j.fitote.2024.105926] [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: 11/29/2023] [Revised: 03/17/2024] [Accepted: 03/24/2024] [Indexed: 04/26/2024]
Abstract
Hyperuricemia (HUA) is a metabolic disease characterized by the increase of serum uric acid (UA) level. Sargentodoxae Caulis (SC) is a commonly used herbal medicine for the treatment of gouty arthritis, traumatic swelling, and rheumatic arthritis in clinic. In this study, a total of fifteen compounds were identified in SC water extract using UHPLC-Q-TOF-MS/MS, including three phenolic acids, seven phenolic glycosides, four organic acids, and one lignan. Then, to study the hypouricemia effect of SC, a HUA mouse model was induced using a combination of PO, HX, and 20% yeast feed. After 14 days of treatment with the SC water extract, the levels of serum UA, creatinine (CRE), blood urea nitrogen (BUN) were reduced significantly, and the organ indexes were restored, the xanthine oxidase (XOD) activity were inhibited as well. Meanwhile, SC water extract could ameliorate the pathological status of kidneys and intestine of HUA mice. Additionally, quantitative real-time PCR (qRT-PCR) and western blotting results showed that SC water extract could increase the expression of ATP binding cassette subfamily G member 2 (ABCG2), organic cation transporter 1 (OCT1), organic anion transporter 1 (OAT1) and organic anion transporter 3 (OAT3), whereas decrease the expression of glucose transporter 9 (GLUT9). This study provided a data support for the clinical application of SC in the treatment of HUA.
Collapse
Affiliation(s)
- Wen-Wen Liu
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Hong-Jing Dong
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Zhe Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Xin-Hui Ma
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Shuang Liu
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Wei Huang
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Xiao Wang
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, China.
| |
Collapse
|
11
|
Yang S, Liu H, Fang XM, Yan F, Zhang Y. Signaling pathways in uric acid homeostasis and gout: From pathogenesis to therapeutic interventions. Int Immunopharmacol 2024; 132:111932. [PMID: 38560961 DOI: 10.1016/j.intimp.2024.111932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/20/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024]
Abstract
Uric acid is a product of purine degradation, and uric acid may have multiple physiologic roles, including the beneficial effects as an antioxidant and neuroprotector, maintenance of blood pressure during low salt ingestion, and modulation of immunity. However, overproduction of metabolic uric acid, and/or imbalance of renal uric acid secretion and reabsorption, and/or underexcretion of extrarenal uric acid, e.g. gut, will contribute to hyperuricemia, which is a common metabolic disease. Long-lasting hyperuricemia can induce the formation and deposition of monosodium urate (MSU) crystals within the joints and periarticular structures. MSU crystals further induce an acute, intensely painful, and sterile inflammation conditions named as gout by NLRP3 inflammasome-mediated cleavage of pro-IL-1β to bioactive IL-1β. Moreover, hyperuricemia and gout are associated with multiple cardiovascular and renal disorders, e.g., hypertension, myocardial infarction, stroke, obesity, hyperlipidemia, type 2 diabetes mellitus and chronic kidney disease. Although great efforts have been made by scientists of modern medicine, however, modern therapeutic strategies with a single target are difficult to exert long-term positive effects, and even some of these agents have severe adverse effects. The Chinese have used the ancient classic prescriptions of traditional Chinese medicine (TCM) to treat metabolic diseases, including gout, by multiple targets, for more than 2200 years. In this review, we discuss the current understanding of urate homeostasis, the pathogenesis of hyperuricemia and gout, and both modern medicine and TCM strategies for this commonly metabolic disorder. We hope these will provide the good references for treating hyperuricemia and gout.
Collapse
Affiliation(s)
- Shuangling Yang
- School of Health Sciences, Guangzhou Xinhua University, Guangzhou, Guangdong 510520, China
| | - Haimei Liu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xian-Ming Fang
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi 530011, China.
| | - Fuman Yan
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Yaxing Zhang
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Issue 12(th) of Guangxi Apprenticeship Education of Traditional Chinese Medicine (Shi‑Cheng Class of Guangxi University of Chinese Medicine), College of Continuing Education, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China.
| |
Collapse
|
12
|
Wen S, Arakawa H, Tamai I. Uric acid in health and disease: From physiological functions to pathogenic mechanisms. Pharmacol Ther 2024; 256:108615. [PMID: 38382882 DOI: 10.1016/j.pharmthera.2024.108615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/02/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024]
Abstract
Owing to renal reabsorption and the loss of uricase activity, uric acid (UA) is strictly maintained at a higher physiological level in humans than in other mammals, which provides a survival advantage during evolution but increases susceptibility to certain diseases such as gout. Although monosodium urate (MSU) crystal precipitation has been detected in different tissues of patients as a trigger for disease, the pathological role of soluble UA remains controversial due to the lack of causality in the clinical setting. Abnormal elevation or reduction of UA levels has been linked to some of pathological status, also known as U-shaped association, implying that the physiological levels of UA regulated by multiple enzymes and transporters are crucial for the maintenance of health. In addition, the protective potential of UA has also been proposed in aging and some diseases. Therefore, the role of UA as a double-edged sword in humans is determined by its physiological or non-physiological levels. In this review, we summarize biosynthesis, membrane transport, and physiological functions of UA. Then, we discuss the pathological involvement of hyperuricemia and hypouricemia as well as the underlying mechanisms by which UA at abnormal levels regulates the onset and progression of diseases. Finally, pharmacological strategies for urate-lowering therapy (ULT) are introduced, and current challenges in UA study and future perspectives are also described.
Collapse
Affiliation(s)
- Shijie Wen
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
13
|
Hussain A, Rui B, Ullah H, Dai P, Ahmad K, Yuan J, Liu Y, Li M. Limosilactobacillus reuteri HCS02-001 Attenuates Hyperuricemia through Gut Microbiota-Dependent Regulation of Uric Acid Biosynthesis and Excretion. Microorganisms 2024; 12:637. [PMID: 38674582 PMCID: PMC11052267 DOI: 10.3390/microorganisms12040637] [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: 01/23/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Hyperuricemia is a prevalent metabolic disorder that arises from abnormal purine metabolism and reduced excretion of uric acid (UA). The gut microbiota plays a significant role in the biosynthesis and excretion of UA. Probiotics capable of purine degradation possess the potential to prevent hyperuricemia. Our study aimed to screen probiotics in areas with abundant dairy products and longevity populations in China, which could attenuate the level of UA and explore the underlying mechanism. In this study, twenty-three lactic acid bacteria isolated from healthy Chinese infant feces and traditional fermented foods such as hurood and lump milk were evaluated for the ability to tolerance acid, bile, artificial gastric juice, and artificial intestinal juice to determine the potential of the candidate strains as probiotics. Eight strains were identified as possessing superior tolerance to simulated intestinal conditions and were further analyzed by high-performance liquid chromatography (HPLC), revealing that Limosilactobacillus reuteri HCS02-001 (Lact-1) and Lacticaseibacillus paracasei HCS17-040 (Lact-2) possess the most potent ability to degrade purine nucleosides. The effect of Lact-1 and Lact-2 on hyperuricemia was evaluated by intervening with them in the potassium oxonate and adenine-induced hyperuricemia Balb/c mice model in vivo. Our results showed that the level of serum UA in hyperuricemic mice can be efficiently reduced via the oral administration of Lact-1 (p < 0.05). It significantly inhibited the levels of liver inflammatory cytokines and hepatic xanthine oxidase through a TLR4/MyD88/NF-κB pathway across the gut-liver axis. Furthermore, UA transporters ABCG2 and SLC2A9 were substantially upregulated by the intervention of this probiotic. Fecal ATP levels were significantly induced, while fecal xanthine dehydrogenase and allantoinase levels were increased following probiotics. RNA sequencing of HT-29 cells line treated with Lact-1 and its metabolites demonstrated significant regulation of pathways related to hyperuricemia. In summary, these findings demonstrate that Limosilactobacillus reuteri HCS02-001 possesses a capacity to ameliorate hyperuricemia by inhibiting UA biosynthesis via enhancing gastrointestinal barrier functions and promoting UA removal through the upregulation of urate transporters, thereby providing a basis for the probiotic formulation by targeting the gut microbiota.
Collapse
Affiliation(s)
- Akbar Hussain
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Binqi Rui
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Hayan Ullah
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Panpan Dai
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Kabir Ahmad
- Department of Physiology, Dalian Medical University, Dalian 116041, China;
| | - Jieli Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| |
Collapse
|
14
|
Fu Y, Chen YS, Xia DY, Luo XD, Luo HT, Pan J, Ma WQ, Li JZ, Mo QY, Tu Q, Li MM, Zhao Y, Li Y, Huang YT, Chen ZX, Li ZJ, Bernard L, Dione M, Zhang YM, Miao K, Chen JY, Zhu SS, Ren J, Zhou LJ, Jiang XZ, Chen J, Lin ZP, Chen JP, Ye H, Cao QY, Zhu YW, Yang L, Wang X, Wang WC. Lactobacillus rhamnosus GG ameliorates hyperuricemia in a novel model. NPJ Biofilms Microbiomes 2024; 10:25. [PMID: 38509085 PMCID: PMC10954633 DOI: 10.1038/s41522-024-00486-9] [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: 09/21/2023] [Accepted: 02/06/2024] [Indexed: 03/22/2024] Open
Abstract
Hyperuricemia (HUA) is a metabolic syndrome caused by abnormal purine metabolism. Although recent studies have noted a relationship between the gut microbiota and gout, whether the microbiota could ameliorate HUA-associated systemic purine metabolism remains unclear. In this study, we constructed a novel model of HUA in geese and investigated the mechanism by which Lactobacillus rhamnosus GG (LGG) could have beneficial effects on HUA. The administration of antibiotics and fecal microbiota transplantation (FMT) experiments were used in this HUA goose model. The effects of LGG and its metabolites on HUA were evaluated in vivo and in vitro. Heterogeneous expression and gene knockout of LGG revealed the mechanism of LGG. Multi-omics analysis revealed that the Lactobacillus genus is associated with changes in purine metabolism in HUA. This study showed that LGG and its metabolites could alleviate HUA through the gut-liver-kidney axis. Whole-genome analysis, heterogeneous expression, and gene knockout of LGG enzymes ABC-type multidrug transport system (ABCT), inosine-uridine nucleoside N-ribohydrolase (iunH), and xanthine permease (pbuX) demonstrated the function of nucleoside degradation in LGG. Multi-omics and a correlation analysis in HUA patients and this goose model revealed that a serum proline deficiency, as well as changes in Collinsella and Lactobacillus, may be associated with the occurrence of HUA. Our findings demonstrated the potential of a goose model of diet-induced HUA, and LGG and proline could be promising therapies for HUA.
Collapse
Affiliation(s)
- Yang Fu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yong-Song Chen
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Dai-Yang Xia
- School of Marine Sciences, Sun Yat-sen University, and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519082, China
| | - Xiao-Dan Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Hao-Tong Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jie Pan
- Hunan Shihua Biotech Co. Ltd., Changsha, 410000, China
| | - Wei-Qing Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jin-Ze Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qian-Yuan Mo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qiang Tu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, China
| | - Meng-Meng Li
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng, 252000, China
| | - Yue Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yi-Teng Huang
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zhi-Xian Chen
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zhen-Jun Li
- Key Laboratory of Carcinogenesis and Translational Research, Departments of Lymphoma, Radiology and Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, 100080, China
| | - Lukuyu Bernard
- International Livestock Research Institute, Nairobi, 00100, Kenya
| | - Michel Dione
- International Livestock Research Institute, Nairobi, 00100, Kenya
| | - You-Ming Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, China
| | - Kai Miao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, SAR, China
| | - Jian-Ying Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shan-Shan Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jie Ren
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Ling-Juan Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xian-Zhi Jiang
- Microbiome Research Center, Moon (Guangzhou) Biotech Co. Ltd., Guangzhou, 510535, China
| | - Juan Chen
- Microbiome Research Center, Moon (Guangzhou) Biotech Co. Ltd., Guangzhou, 510535, China
| | - Zhen-Ping Lin
- Shantou Baisha Research Institute of Origin Species of Poultry and Stock, Shantou, 515041, China
| | - Jun-Peng Chen
- Shantou Baisha Research Institute of Origin Species of Poultry and Stock, Shantou, 515041, China
| | - Hui Ye
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qing-Yun Cao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yong-Wen Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Lin Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Xue Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, China.
| | - Wen-Ce Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
15
|
Yu Y, Yang X, Hu G, Tong K, Yin Y, Yu R. Effect of tea intake on genetic predisposition to gout and uric acid: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 14:1290731. [PMID: 38440060 PMCID: PMC10911082 DOI: 10.3389/fendo.2023.1290731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/26/2023] [Indexed: 03/06/2024] Open
Abstract
Objective The effect of tea on gout and uric acid is still controversial. This study aims to analyze the effect of tea intake on genetic predisposition to gout, idiopathic gout, gout due to impairment of renal function as well as uric acid by Mendelian randomization (MR). Methods Forty independent single nucleotide polymorphisms (SNPs) associated with tea intake were selected from UK Biobank. SNPs for uric acid were obtained from BioBank Japan, SNPs for gout were obtained from UK Biobank, and SNPs for gout due to impairment of renal function and idiopathic gout were derived from FinnGen. The causal relationship of exposure-outcome was tested using inverse variance weighted, MR-Egger and weighted median. MR-Egger intercept was employed to assess horizontal pleiotropy, Cochran's Q test was used to assess heterogeneity, and leave-one-out sensitivity analysis was utilized to analyze the stability of the results. Results The results of MR analysis showed that tea intake was negatively associated with gout due to impairment of renal function (OR 0.997, 95% CI 0.994 to 0.999, P = 0.017), whereas there was no causal association with gout, idiopathic gout, and uric acid (P > 0.05), for which sensitivity analysis suggested that these results were robust. Conclusions There was a genetic predisposition effect of increased tea intake on the reduced risk of gout due to impairment of renal function, whereas there was no such effect on gout, idiopathic gout, and uric acid. Tea intake may become an important option in the dietary treatment of gout due to impairment of renal function.
Collapse
Affiliation(s)
- Yunfeng Yu
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyu Yang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gang Hu
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Keke Tong
- Department of Gastroenterology, The Hospital of Hunan University of Traditional Chinese Medicine, Changde, Hunan, China
| | - Yuman Yin
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rong Yu
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
16
|
Wang R, Sun Y, Wang H, Liu T, Shavandi A, Nie L, Yunusov KE, Jiang G. Core-shell structured microneedles with programmed drug release functions for prolonged hyperuricemia management. J Mater Chem B 2024; 12:1064-1076. [PMID: 38168723 DOI: 10.1039/d3tb02607h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
An appropriate non-oral platform via transdermal delivery of drugs is highly recommended for the treatment of hyperuricemia. Herein, a core-shell structured microneedle patch with programmed drug release functions was designed to regulate serum uric acid (SUA) levels for prolonged hyperuricemia management. The patch was fabricated using a three-step casting method. Allopurinol (AP), an anti-hyperuricemic drug, was encapsulated within the carboxymethyl cellulose (CMC) layer, forming the "shell" of the MNs. The MN's inner core was composed of polyvinylpyrrolidone (PVP) loaded with urate oxidase-calcium peroxide nanoparticles (UOx-CaO2 NPs). When the as-fabricated core-shell structured microneedles were inserted into the skin, the loaded AP was first released immediately to effectively inhibit the production of SUA due to the water solubility of CMC. Subsequently, the internal SUA was further metabolized by UOx, leading to exposure of CaO2 NPs. The sustained release of UOx accompanied by the decomposition of CaO2 NPs contributed to maintaining a state of normal uric acid levels over an extended period. More attractively, uric acid could be oxidized due to the strong oxidant of CaO2, which was beneficial to the continuous consumption of uric acid. In vivo results showed that the as-fabricated MNs exhibited an excellent anti-hyperuricemia effect to reduce SUA levels to the normal state within 3 h and maintain the normouricemia state for 12 h. In addition, the levels of creatinine (Cr) and blood urea nitrogen (BUN) in the serum remained within the normal range, and the activities of adenosine deaminase (ADA) and xanthine oxidase (XOD) in the liver were effectively inhabited, mitigating the risk of liver and kidney damage for clinical anti-hyperuricemia management.
Collapse
Affiliation(s)
- Rui Wang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, China
| | - Yanfang Sun
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China.
| | - Han Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Tianqi Liu
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, China
| | - Amin Shavandi
- BioMatter unit-École polytechnique de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Lei Nie
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Khaydar E Yunusov
- Institute of Polymer Chemistry and Physics, Uzbekistan Academy of Sciences, Tashkent, 100128, Uzbekistan
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers of Zhejiang Province, Hangzhou, 310018, China
| |
Collapse
|
17
|
Wang M, Bai QX, Zheng XX, Hu WJ, Wang S, Tang HP, Yu AQ, Yang BY, Kuang HX. Smilax china L.: A review of its botany, ethnopharmacology, phytochemistry, pharmacological activities, actual and potential applications. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116992. [PMID: 37541403 DOI: 10.1016/j.jep.2023.116992] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Smilax china L., an extensively used traditional Chinese medicine, is known as Baqia in China. It has been used to treat various inflammatory disorders, particularly pelvic inflammation. AIM OF THE REVIEW The present paper aims to provide an up-to-date review at the advancements of the investigations on the ethnopharmacology, phytochemistry, pharmacological effect and actual and potential applications of S. china. Besides, the possible tendency and perspective for future research of this plant are discussed, as well. MATERIALS AND METHODS This article uses "Smilax china L." "S. china" as the keyword and collects relevant information on Smilax china L. plants through electronic searches (Elsevier, PubMed, ACS, CNKI, Google Scholar, Baidu Scholar, Web of Science), relevant books, and classic literature about Chinese herb. RESULTS 134 chemical constituents, among which steroid saponins and flavonoids are the predominant groups, have been isolated and identified from S. china. S. china with its active compounds is possessed of wide-reaching biological activities, including anti-inflammatory, anti-cancer, anti-oxidant, detoxify nicotine, anti-diabetes, anti-obesity, anti-hyperuricaemia, anti-hypertension, promoting skin wound and barrier repair and anti-bacterial activity. Besides, S. china is also applied to other fields, such as food industry and detection technology. CONCLUSIONS Based on the review of the existing phytochemical studies on Smilax china L., the structural characterization of Smilax china L. extract can continue to be the focus of future research. Pharmacological studies in vitro and in vivo have demonstrated some of the traditional uses of Smilax china L. extract, while other traditional uses still need to be confirmed by research.
Collapse
Affiliation(s)
- Meng Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China.
| | - Qian-Xiang Bai
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Xiu-Xi Zheng
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Wen-Jing Hu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Shuang Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Hai-Peng Tang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Ai-Qi Yu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Bing-You Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Hai-Xue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, 150000, China.
| |
Collapse
|
18
|
Ma N, Cai S, Sun Y, Chu C. Chinese Sumac ( Rhus chinensis Mill.) Fruits Prevent Hyperuricemia and Uric Acid Nephropathy in Mice Fed a High-Purine Yeast Diet. Nutrients 2024; 16:184. [PMID: 38257077 PMCID: PMC10819650 DOI: 10.3390/nu16020184] [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: 11/07/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Hyperuricemia (HUA) is a prevalent chronic disease, characterized by excessive blood uric acid levels, that poses a significant health risk. In this study, the preventive effects and potential mechanisms of ethanol extracts from Chinese sumac (Rhus chinensis Mill.) fruits on HUA and uric acid nephropathy were comprehensively investigated. The results demonstrated a significant reduction in uric acid levels in hyperuricemia mice after treatment with Chinese sumac fruit extract, especially in the high-dose group, where the blood uric acid level decreased by 39.56%. Visual diagrams of the kidneys and hematoxylin and eosin (H&E)-stained sections showed the extract's effectiveness in protecting against kidney damage caused by excessive uric acid. Further investigation into its mechanism revealed that the extract prevents and treats hyperuricemia by decreasing uric acid production, enhancing uric acid excretion, and mitigating the oxidative stress and inflammatory reactions induced by excessive uric acid in the kidneys. Specifically, the extract markedly decreased xanthine oxidase (XOD) levels and expression in the liver, elevated the expression of uric acid transporters ABCG2, and lowered the expression of uric acid reabsorption proteins URAT1 and SLC2A9. Simultaneously, it significantly elevated the levels of endogenous antioxidant enzymes (SOD and GSH) while reducing the level of malondialdehyde (MDA). Furthermore, the expression of uric-acid-related proteins NLRP3, ACS, and Caspase-3 and the levels of IL-1β and IL-6 were significantly reduced. The experimental results confirm that Chinese sumac fruit extract can improve HUA and uric acid nephropathy in mice fed a high-purine yeast diet. This finding establishes a theoretical foundation for developing Chinese sumac fruit as a functional food or medicine for preventing and treating HUA.
Collapse
Affiliation(s)
| | | | | | - Chuanqi Chu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (N.M.); (S.C.)
| |
Collapse
|
19
|
Tang Y, Du Y, Ye J, Deng L, Cui W. Intestine-Targeted Explosive Hydrogel Microsphere Promotes Uric Acid Excretion for Gout Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310492. [PMID: 37997010 DOI: 10.1002/adma.202310492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/04/2023] [Indexed: 11/25/2023]
Abstract
Uric acid metabolism disorder triggers metabolic diseases, especially gout. However, increasing uric acid excretion remains a challenge. Here, an accelerative uric acid excretion pathway via an oral intestine-explosive hydrogel microsphere merely containing uricase and dopamine is reported. After oral administration, uricase is exposed and immobilized on intestinal mucosa along with an in situ dopamine polymerization via a cascade reaction triggered by the intestinal specific environment. By this means, trace amount of uricase is required to in situ up-regulate uric acid transporter proteins of intestinal epithelial cells, causing accelerated intestinal uric acid excretion. From in vitro data, the uric acid in fecal samples from gout patients could be significantly reduced by up to 37% by the mimic mucosa-immobilized uricase on the isolated porcine tissues. Both hyperuricemia and acute gouty arthritis in vivo mouse models confirm the uric acid excretion efficacy of intestine-explosive hydrogel microspheres. Fecal uric acid excretion is increased around 30% and blood uric acid is reduced more than 70%. In addition, 16S ribosomal RNA sequencing showed that the microspheres optimized intestinal flora composition as well. In conclusion, a unique pathway via the intestine in situ regulation to realize an efficient uric acid intestinal excretion for gout therapy is developed.
Collapse
Affiliation(s)
- Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yawei Du
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Junna Ye
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
20
|
Wang J, Hao P, Sun X, Ward R, Tang T, Chen X, Liu Y, Luo G, Yang Y, Xiang C, An S, Xu TR. New animal model of chronic gout reproduces pathological features of the disease in humans. RMD Open 2023; 9:e003499. [PMID: 37973536 PMCID: PMC10660916 DOI: 10.1136/rmdopen-2023-003499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023] Open
Abstract
OBJECTIVES Gout, as the most prevalent form of inflammatory arthritis, necessitates the use of animal models to investigate the molecular mechanisms involved in its development. Therefore, our objective was to develop a novel chronic mouse model of gout that more closely mimics the progression of gout in humans. METHODS A novel chronic mouse model of gout was established by a simple method, which does not require high technical proficiency, predominantly involves daily intraperitoneal injections of potassium oxonate for approximately 4 months, combined with a high fat-diet and injections of acetic acid into the hind paws to facilitate the formation of monosodium urate (MSU). Arthritis scores and paw oedema were assessed, behavioural tests were conducted, and histopathological and imaging evaluations of the arthritic paw joints were performed. RESULTS After 4 months of induction, mice in the model group exhibited noticeable increases in arthritis severity, joint and cartilage damage, as well as bone erosion. Gomori's methenamine silver stain revealed the presence of MSU crystal deposition or tophi in the paw joints or ankle joints of up to 37.9% of the model mice (11 out of 29 mice). Moreover, treatment with benzbromarone effectively prevented the further development of gout or tophi formation in model mice. CONCLUSIONS Our model more accurately replicates the pathological features of gouty arthritis compared with gout induced by MSU crystal injections. Therefore, it is particularly suitable for further investigations into the pathogenesis of gout and also serves as a valuable platform for screening potential antigout agents.
Collapse
Affiliation(s)
- Jiwei Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Peiqi Hao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xianrun Sun
- Department of Orthopedic Surgery, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Richard Ward
- Centre for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Tao Tang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xi Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yihong Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Guancong Luo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Cheng Xiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Su An
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
21
|
Liu Y, Jarman JB, Low YS, Augustijn HE, Huang S, Chen H, DeFeo ME, Sekiba K, Hou BH, Meng X, Weakley AM, Cabrera AV, Zhou Z, van Wezel G, Medema MH, Ganesan C, Pao AC, Gombar S, Dodd D. A widely distributed gene cluster compensates for uricase loss in hominids. Cell 2023; 186:3400-3413.e20. [PMID: 37541197 PMCID: PMC10421625 DOI: 10.1016/j.cell.2023.06.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 03/22/2023] [Accepted: 06/19/2023] [Indexed: 08/06/2023]
Abstract
Approximately 15% of US adults have circulating levels of uric acid above its solubility limit, which is causally linked to the disease gout. In most mammals, uric acid elimination is facilitated by the enzyme uricase. However, human uricase is a pseudogene, having been inactivated early in hominid evolution. Though it has long been known that uric acid is eliminated in the gut, the role of the gut microbiota in hyperuricemia has not been studied. Here, we identify a widely distributed bacterial gene cluster that encodes a pathway for uric acid degradation. Stable isotope tracing demonstrates that gut bacteria metabolize uric acid to xanthine or short chain fatty acids. Ablation of the microbiota in uricase-deficient mice causes severe hyperuricemia, and anaerobe-targeted antibiotics increase the risk of gout in humans. These data reveal a role for the gut microbiota in uric acid excretion and highlight the potential for microbiome-targeted therapeutics in hyperuricemia.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - J Bryce Jarman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Hannah E Augustijn
- Bioinformatics Group, Wageningen University, Wageningen, the Netherlands; Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Steven Huang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Haoqing Chen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mary E DeFeo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kazuma Sekiba
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bi-Huei Hou
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiandong Meng
- ChEM-H Institute, Stanford University, Stanford, CA 94305, USA
| | | | | | - Zhiwei Zhou
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gilles van Wezel
- Institute of Biology, Leiden University, Leiden, the Netherlands; Netherlands Institute of Ecology, Wageningen, the Netherlands
| | - Marnix H Medema
- Bioinformatics Group, Wageningen University, Wageningen, the Netherlands; Institute of Biology, Leiden University, Leiden, the Netherlands
| | - Calyani Ganesan
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan C Pao
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Urology, Stanford University School of Medicine, Stanford, CA 94305, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Saurabh Gombar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Atropos Health, Palo Alto, CA, USA
| | - Dylan Dodd
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Sun L, Liu Q, Zhang Y, Xue M, Yan H, Qiu X, Tian Y, Zhang H, Liang H. Fucoidan from Saccharina japonica Alleviates Hyperuricemia-Induced Renal Fibrosis through Inhibiting the JAK2/STAT3 Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:11454-11465. [PMID: 37481747 DOI: 10.1021/acs.jafc.3c01349] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Fucoidan is a native sulfated polysaccharide mainly isolated from brown seaweed, with diverse pharmacological activities, such as anti-inflammatory and antifibrosis. Hyperuricemia (HUA) is a common metabolic disease worldwide and mainly causes hyperuricemic nephropathy, including chronic kidney disease and end-stage renal fibrosis. The present study investigated the protective function of fucoidan in renal fibrosis and its pharmacological mechanism. The renal fibrotic model was established with the administration of potassium oxonate for 10 weeks. The protein levels of related factors were assessed in HUA mice by an enzyme-linked immunosorbent assay (ELISA) and western blotting. The results showed that fucoidan significantly reduced the levels of serum uric acid, blood urea nitrogen (BUN), α-smooth muscle actin (α-SMA), and collagen I, and improved kidney pathological changes. Furthermore, renal fibrosis had been remarkably elevated through the inhibition of the epithelial-to-mesenchymal transition (EMT) progression after fucoidan intervention, suppressing the Janus kinase 2 (JAK2) signal transducer and activator of transcription protein 3 (STAT3) signaling pathway activation. Together, this study provides experimental evidence that fucoidan may protect against hyperuricemia-induced renal fibrosis via downregulation of the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Lirui Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Qing Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Yabin Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Meilan Xue
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Hongxue Yan
- State Key Laboratory of Bioactive Seaweed Substances, Qingdao Mingyue Seaweed Group Company, Limited, Qingdao, Shandong 266499, People's Republic of China
| | - Xia Qiu
- State Key Laboratory of Bioactive Seaweed Substances, Qingdao Mingyue Seaweed Group Company, Limited, Qingdao, Shandong 266499, People's Republic of China
| | - Yingjie Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Huaqi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Hui Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| |
Collapse
|
23
|
Li Y, Pu LY, Li Y, Zhu G, Wu Z. Design, synthesis and evaluation of a myricetin and nobiletin hybrid compound for alleviating hyperuricemia based on metabolomics and gut microbiota. RSC Adv 2023; 13:21448-21458. [PMID: 37465570 PMCID: PMC10351364 DOI: 10.1039/d3ra03188h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/01/2023] [Indexed: 07/20/2023] Open
Abstract
Hyperuricemia (HUA) is the fourth most common basic metabolic disease that can cause damage to multiple organs throughout the body. In this study, a hybrid compound consisting of myricetin and nobiletin was synthesized and its biological activity was evaluated. We named the hybrid compound MNH, and its structure was confirmed by spectroscopy. This study used serum metabolomics profiling with LC/MS and 16S rRNA gene sequencing analysis to explore the anti-HUA efficacy of MNH on a yeast paste-induced mouse model. The results showed that serum uric acid (UA), creatinine (CRE) and urea nitrogen (BUN) levels were significantly decreased after the intervention of MNH. The efficacy of MNH in lowering UA was somewhat greater than that of myricetin and nobiletin. In addition, MNH could repair the renal histopathological damage. Moreover, serum metabolomics demonstrated that MNH regulated the metabolic pathways involved in glycerophospholipid metabolism, arachidonic acid metabolism and alanine etc. Furthermore, MNH supplementation restored the composition of gut microbiota with remarkable reductions in Lactobacillus and Limosilactobacillus and significant elevations in norank_f_Muribaculaceae and Bacteroides at the genus level. Taken together, these results indicated that MNH might represent a protective effect against HUA via modulating gut microbiota and metabolomics.
Collapse
Affiliation(s)
- Yan Li
- Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital Shenzhen 518035 China
- Wu Zhengzhi Academician Workstation, NingBo College of Health Sciences Ningbo 315800 China
- Shenzhen Institute of Geriatrics Shenzhen 518035 China
| | - Liu-Yang Pu
- Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital Shenzhen 518035 China
- Shenzhen Institute of Geriatrics Shenzhen 518035 China
| | - Yayun Li
- Hunan University of Chinese Medicine Changsha 410208 China
| | - Guanbao Zhu
- Guangxi University of Chinese Medicine Nanning 530200 China
| | - Zhengzhi Wu
- Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital Shenzhen 518035 China
- Wu Zhengzhi Academician Workstation, NingBo College of Health Sciences Ningbo 315800 China
- Shenzhen Institute of Geriatrics Shenzhen 518035 China
| |
Collapse
|
24
|
Rodríguez-Rovira I, López-Sainz A, Palomo-Buitrago ME, Pérez B, Jiménez-Altayó F, Campuzano V, Egea G. Hyperuricaemia Does Not Interfere with Aortopathy in a Murine Model of Marfan Syndrome. Int J Mol Sci 2023; 24:11293. [PMID: 37511051 PMCID: PMC10379183 DOI: 10.3390/ijms241411293] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Redox stress is involved in the aortic aneurysm pathogenesis in Marfan syndrome (MFS). We recently reported that allopurinol, a xanthine oxidoreductase inhibitor, blocked aortopathy in a MFS mouse model acting as an antioxidant without altering uric acid (UA) plasma levels. Hyperuricaemia is ambiguously associated with cardiovascular injuries as UA, having antioxidant or pro-oxidant properties depending on the concentration and accumulation site. We aimed to evaluate whether hyperuricaemia causes harm or relief in MFS aortopathy pathogenesis. Two-month-old male wild-type (WT) and MFS mice (Fbn1C1041G/+) were injected intraperitoneally for several weeks with potassium oxonate (PO), an inhibitor of uricase (an enzyme that catabolises UA to allantoin). Plasma UA and allantoin levels were measured via several techniques, aortic root diameter and cardiac parameters by ultrasonography, aortic wall structure by histopathology, and pNRF2 and 3-NT levels by immunofluorescence. PO induced a significant increase in UA in blood plasma both in WT and MFS mice, reaching a peak at three and four months of age but decaying at six months. Hyperuricaemic MFS mice showed no change in the characteristic aortic aneurysm progression or aortic wall disarray evidenced by large elastic laminae ruptures. There were no changes in cardiac parameters or the redox stress-induced nuclear translocation of pNRF2 in the aortic tunica media. Altogether, the results suggest that hyperuricaemia interferes neither with aortopathy nor cardiopathy in MFS mice.
Collapse
Affiliation(s)
- Isaac Rodríguez-Rovira
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Angela López-Sainz
- Department of Cardiology, Hospital Clínic de Barcelona, IDIBAPS, 08036 Barcelona, Spain
| | | | - Belen Pérez
- Department of Pharmacology, School of Medicine, Autonomous University of Barcelona, Bellaterra, 08192 Barcelona, Spain
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, School of Medicine, Autonomous University of Barcelona, Bellaterra, 08192 Barcelona, Spain
| | - Victoria Campuzano
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- UZA/UA Center of Medical Genetics, University of Antwerp, 2650 Edegem, Belgium
| |
Collapse
|
25
|
Pei XD, Li F, Zhang YM, Huang XN, Yu FT, Su LY, Liu XL, Wang CH. Preparation, Purification, and Identification of Novel Feather Keratin-Derived Peptides with Antioxidative and Xanthine Oxidase Inhibitory Activities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:8061-8070. [PMID: 37161263 DOI: 10.1021/acs.jafc.3c01131] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Feather keratin is an underappreciated protein resource of high quality, with limited bioavailability, and it urgently requires eco-friendly methods to enhance its value. Here, we report on the preparation, purification, and identification of novel peptides with antioxidant and xanthine oxidase (XOD) inhibitory activities from fermented feather broth, using Bacillus licheniformis 8-4. Two peptides, namely, DLCRPCGPTPLA (DA-12) and ANSCNEPCVR (AR-10), displayed remarkable 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical and 2,2'-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid) (ABTS) radical scavenging abilities with half-maximal inhibitory concentrations (IC50) values of 0.048, 0.034, and 0.95, 0.84 mg/mL, respectively. These values exceed those of the previously reported feather keratin-derived antioxidant peptides. Another peptide, GNQQVHLQSQDM (GM-12), demonstrated XOD activity inhibition, with an IC50 value of 12.15 mg/mL, and it quenched the fluorescence of XOD. Furthermore, after simulating gastrointestinal digestion, DA-12, AR-10, and GM-12 retained their biological activities. Meanwhile, DA-12 and GM-12 showed an unexpected synergistic inhibition on XOD activity accompanied by fluorescence quenching. This study provides new insights into the potential applications of feather keratin, including functionalized feed with antioxidative and antigout (anti-hyperuricemia) activities.
Collapse
Affiliation(s)
- Xiao-Dong Pei
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, People's Republic of China
| | - Fan Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, People's Republic of China
| | - Yan-Mei Zhang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, People's Republic of China
| | - Xiao-Ni Huang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, People's Republic of China
| | - Fu-Tian Yu
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, People's Republic of China
| | - Lin-Ying Su
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, People's Republic of China
| | - Xiao-Ling Liu
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, People's Republic of China
| | - Cheng-Hua Wang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, People's Republic of China
| |
Collapse
|
26
|
Cao J, Wang T, Liu Y, Zhou W, Hao H, Liu Q, Yin B, Yi H. Lactobacillus fermentum F40-4 ameliorates hyperuricemia by modulating the gut microbiota and alleviating inflammation in mice. Food Funct 2023; 14:3259-3268. [PMID: 36928268 DOI: 10.1039/d2fo03701g] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Hyperuricemia (HUA) is a systemic disease characterized by a disorder of purine metabolism and an abnormal increase in the serum level of uric acid (UA). Probiotics can exert potential therapeutic benefits against some metabolic diseases by regulating the intestinal microbiota. Lactobacillus fermentum F40-4 with UA-lowering activity of 87.40% was screened using purine as the target in vitro. The UA-lowering activity of L. fermentum F40-4 was further explored in a mouse model of HUA in vivo. L. fermentum F40-4 could downregulate serum levels of UA, blood urea nitrogen, creatinine, and xanthine oxidase by 40.84%, 11.61%, 57.66%, and 41.79%, respectively. L. fermentum F40-4 restored organ damage, and adjusted enzyme activity and transporter expression to promote the metabolic level of UA. In addition, L. fermentum F40-4 could reshape the gut microbiota and suppress inflammation to ameliorate HUA. An increment in intestinal UA excretion was documented. These findings suggest that L. fermentum F40-4 might serve as a potential probiotic for the prevention and treatment of HUA.
Collapse
Affiliation(s)
- Jiayuan Cao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266000, China.
| | - Ting Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266000, China.
| | - Yisuo Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266000, China.
| | - Wei Zhou
- Yangzhou University Healthy source Dairy Co., LTD., Yangzhou, 225002, China.
| | - Haining Hao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266000, China.
| | - Qiqi Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266000, China.
| | - Boxing Yin
- Yangzhou University Healthy source Dairy Co., LTD., Yangzhou, 225002, China.
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266000, China.
| |
Collapse
|
27
|
Yu W, Xie D, Yamamoto T, Koyama H, Cheng J. Mechanistic insights of soluble uric acid-induced insulin resistance: Insulin signaling and beyond. Rev Endocr Metab Disord 2023; 24:327-343. [PMID: 36715824 DOI: 10.1007/s11154-023-09787-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/31/2023]
Abstract
Hyperuricemia is a metabolic disease caused by purine nucleotide metabolism disorder. The prevalence of hyperuricemia is increasing worldwide, with a growing trend in the younger populations. Although numerous studies have indicated that hyperuricemia may be an independent risk factor for insulin resistance, the causal relationship between the two is controversial. There are few reviews, however, focusing on the relationship between uric acid (UA) and insulin resistance from experimental studies. In this review, we summarized the experimental models related to soluble UA-induced insulin resistance in pancreas and peripheral tissues, including skeletal muscles, adipose tissue, liver, heart/cardiomyocytes, vascular endothelial cells and macrophages. In addition, we summarized the research advances about the key mechanism of UA-induced insulin resistance. Moreover, we attempt to identify novel targets for the treatment of hyperuricemia-related insulin resistance. Lastly, we hope that the present review will encourage further researches to solve the chicken-and-egg dilemma between UA and insulin resistance, and provide strategies for the pathogenesis and treatment of hyperuricemia related metabolic diseases.
Collapse
Affiliation(s)
- Wei Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - De Xie
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Tetsuya Yamamoto
- Health Evaluation Center, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, Nishinomiya, Hyogo, Japan.
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, Fujian, China.
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, People's Republic of China.
| |
Collapse
|
28
|
Wei WH, Guo H. Leveraging primate-specific genomic information for genetic studies of complex diseases. FRONTIERS IN BIOINFORMATICS 2023; 3:1161167. [PMID: 37056664 PMCID: PMC10086602 DOI: 10.3389/fbinf.2023.1161167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Genomic changes specific to higher primates are regarded as primate-specific genomic information (PSI). Using PSI to inform genetic studies is highly desirable but hampered by three factors: heterogeneity among PSI studies, lack of integrated profiles of the identified PSI elements and dearth of relevant functional information. We report a database of 19,767 PSI elements collated from nine types of brain-related studies, which form 19,473 non-overlapping PSI regions that distribute unevenly but jointly cover only 0.81% of the genome. About 2.5% of the PSI regions colocalized with variants identified in genome-wide association studies, with disease loci more likely colocalized than quantitative trait loci (p = 1.6 × 10−5), particularly in regions without obvious regulatory roles. We further showed an LRP4 exemplar region with PSI elements orchestrated with common and rare disease variants and other functional elements. Our results render PSI elements as a valuable source to inform genetic studies of complex diseases.
Collapse
Affiliation(s)
- Wen-Hua Wei
- Department of Women’s and Children’s Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Centre for Biostatistics, School of Health Sciences, The University of Manchester, Manchester, United Kingdom
- *Correspondence: Wen-Hua Wei,
| | - Hui Guo
- Centre for Biostatistics, School of Health Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
Wang R, Wang H, Jiang G, Sun Y, Liu T, Nie L, Shavandi A, Yunusov KE, Aharodnikau UE, Solomevich SO. Transdermal delivery of allopurinol to acute hyperuricemic mice via polymer microneedles for the regulation of serum uric acid levels. Biomater Sci 2023; 11:1704-1713. [PMID: 36628631 DOI: 10.1039/d2bm01836e] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Allopurinol (AP) is widely used to treat hyperuricemia which may cause severe side effects upon oral administration. Alternative means for the treatment of hyperuricemia are demanded to simultaneously facilitate drug absorption, patient compliance, and fewer side effects. In this study, a new polymer microneedle (MN) system was developed for the transdermal delivery of AP to acute hyperuricemic mice. This study aims to achieve the controllable regulation of serum uric acid (SUA) levels with fewer side effects compared with oral administration. The matrix of polymer MNs consisted of polyvinylpyrrolidone (PVP) and polycaprolactone (PCL), in which the rapid dissolution of PVP offers a rapid dissolution of AP into the blood and the biodegradability of PCL resulting in a sustainable drug release behavior. An in vivo study demonstrated that the AP-loaded MN system can effectively reduce the SUA levels as oral administration with lower side effects, which will be conducive to reducing the adverse reactions and improving the bioavailability of AP. This MN-mediated strategy can facilitate transcutaneous hyperuricemia treatment and provide a new alternative for the exploration of clinical treatment of hyperuricemia and improvement of patient compliance.
Collapse
Affiliation(s)
- Rui Wang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Han Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China. .,International Scientific and Technological Cooperation Base of Intelligent Biomaterials and Functional Fibers, Hangzhou, 310018, China
| | - Yanfang Sun
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China.
| | - Tianqi Liu
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Lei Nie
- College of Life Science, Xinyang Normal University, Xinyang, China.,3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Amin Shavandi
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Khaydar E Yunusov
- Institute of Polymer Chemistry and Physics, Uzbekistan Academy of Sciences, Tashkent, Uzbekistan
| | | | - Sergey O Solomevich
- Research Institute for Physical Chemical Problems, Belarusian State University, Minsk, Belarus
| |
Collapse
|
30
|
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
|
31
|
Song D, Zhao H, Wang L, Wang F, Fang L, Zhao X. Ethanol extract of Sophora japonica flower bud, an effective potential dietary supplement for the treatment of hyperuricemia. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
32
|
Platinum nanoparticles confined in metal-organic frameworks as excellent peroxidase-like nanozymes for detection of uric acid. Anal Bioanal Chem 2023; 415:649-658. [PMID: 36443450 DOI: 10.1007/s00216-022-04453-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Abstract
High levels of uric acid (UA) in humans can cause a range of diseases, and traditional assays that rely on uric acid enzymes to break down uric acid are limited by the inherent deficiencies of natural enzymes. Fortunately, the rapid development of nanozymes in recent years is expected to solve the above-mentioned problems. Hence, we used a host-guest strategy to synthesize a platinum nanoparticle confined in a metal-organic framework (Pt NPs@ZIF) that can sensitively detect UA levels in human serum. Unlike previously reported free radical-catalyzed oxidation systems, its unique electron transfer mechanism confers excellent peroxidase-like activity to Pt NPs@ZIF. In addition, UA can selectively inhibit the chromogenic reaction of TMB, thus reducing the absorbance of the system. Therefore, using the peroxidase-like activity of Pt NPs@ZIF and using TMB as a chromogenic substrate, UA can be detected directly without relying on natural enzymes. The results showed a relatively wide detection range (10-1000 μM) and a low detection limit (0.2 μM). Satisfactory results were also obtained for UA in human serum. This study with simple operation and rapid detection offers a promising method for efficiently detecting UA in serum.
Collapse
|
33
|
Yang B, Xin M, Liang S, Huang Y, Li J, Wang C, Liu C, Song X, Sun J, Sun W. Naringenin Ameliorates Hyperuricemia by Regulating Renal Uric Acid Excretion via the PI3K/AKT Signaling Pathway and Renal Inflammation through the NF-κB Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1434-1446. [PMID: 36525382 DOI: 10.1021/acs.jafc.2c01513] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Hyperuricemia characterized by high serum levels of uric acid (UA, >6.8 mg/dL) is regarded as a common chronic metabolic disease. When used as a food supplement, naringenin might have various pharmacological activities, including antioxidant, free-radical-scavenging, and inflammation-suppressing activities. However, the effects of naringenin on hyperuricemia and renal inflammation and the underlying mechanisms remain to be elucidated. Here, we comprehensively examined the effects of naringenin on hyperuricemia and the attenuation of renal impairment. Mice were injected with 250 mg/kg of potassium oxonate (PO) and given 5% fructose water to induce hyperuricemia. The pharmacological effects of naringenin (10 and 50 mg/kg) and benzbromarone (positive control group, 20 mg/kg) on hyperuricemic mice were evaluated in vivo. The disordered expression of urate transporters in HK-2 cells was stimulated by 8 mg/dL UA, which was used to determine the mechanisms underlying the effects of naringenin in vitro. Naringenin markedly reduced the serum UA level in a dose-dependent manner and improved renal dysfunction. Moreover, the increased elimination of UA in urine showed that the effects of naringenin were associated with the regulation of renal excretion. Further examination indicated that naringenin reduced the expression of GLUT9 by inhibiting the PI3K/AKT signaling pathway and reinforced the expression of ABCG2 by increasing the abundance of PDZK1 in vivo and in vitro. Furthermore, sirius red staining and western blotting indicated that naringenin plays a protective role in renal injury by suppressing increases in the levels of pro-inflammatory cytokines, including IL-6 and TNF-α, which contribute to the inhibition of the TLR4/NF-κB signaling pathway in vivo and in vitro. Naringenin supplementation might be a potential therapeutic strategy to ameliorate hyperuricemia by promoting UA excretion in the kidney and attenuating the inflammatory response by decreasing the release of inflammatory cytokines. This study shows that naringenin could be used as a functional food or dietary supplement for hyperuricemia prevention and treatment.
Collapse
Affiliation(s)
- Bendong Yang
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
| | - Meiling Xin
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
| | - Shufei Liang
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
| | - Yuhong Huang
- College of Life Science, Yangtze University, Jingzhou, Hubei 434100, People's Republic of China
| | - Jingda Li
- College of Life Science, Yangtze University, Jingzhou, Hubei 434100, People's Republic of China
| | - Chao Wang
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, Shandong 250100, People's Republic of China
| | - Xinhua Song
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
- Shandong Qingyujiangxing Biotechnology Company, Limited, Zibo, Shandong 255000, People's Republic of China
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, Shandong 250100, People's Republic of China
- School of Public Health and Management, Weifang Medical University, 7166 Baotong Road, Weifang, Shandong 261053, People's Republic of China
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, People's Republic of China
- Shandong Qingyujiangxing Biotechnology Company, Limited, Zibo, Shandong 255000, People's Republic of China
| |
Collapse
|
34
|
Yang Y, Hu Y, Yao F, Yang J, Ge L, Wang P, Xu X. Virtual screening and activity evaluation of human uric acid transporter 1 (hURAT1) inhibitors. RSC Adv 2023; 13:3474-3486. [PMID: 36756549 PMCID: PMC9871872 DOI: 10.1039/d2ra07193b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/13/2022] [Indexed: 01/25/2023] Open
Abstract
Hyperuricemia is a disease caused by disorder of purine metabolism, mainly due to insufficient renal excretion of uric acid. Urate transporter 1 (URAT1) is the most widely studied target of urate transporters, and used for uric acid (UA) reabsorption. This study used the AlphaFold2 algorithm to predict the structure of URAT1. Virtual screening and biological evaluation were used to discover novel URAT1 inhibitors that target the critical amino acids. Seven compounds were screened from the T2220 database and validated as URAT1 inhibitors by cell biology experiments. The IC50 values of benbromarone, NP023335, TN1148, and TN1008 were 6.878, 18.46, 24.64, and 53.04 μM, respectively. Molecular dynamics simulation was used to investigate the binding mechanism of URAT1 to NP023335, which forms stable contact with Ser35, Phe365, and Arg477. These interactions are essential for maintaining the biological activity of NP023335. The three compounds' pharmacokinetic characteristics were predicted, and NP023335's properties matched those of an empirical medication with the benefits of high solubility, low cardiotoxicity, good membrane permeability, and oral absorption. The natural product NP023335 will serve as a promising hit compound for facilitating the further design of novel URAT1 inhibitors.
Collapse
Affiliation(s)
- Yacong Yang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
- Marine Drug Screening and Evaluation Platform (QNLM), Ocean University of China Qingdao 266071 China
| | - Yu Hu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
- Marine Drug Screening and Evaluation Platform (QNLM), Ocean University of China Qingdao 266071 China
| | - Fengli Yao
- College of Food Science and Engineering, Ocean University of China Qingdao 266071 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
- Marine Drug Screening and Evaluation Platform (QNLM), Ocean University of China Qingdao 266071 China
- School of Life Science, Lanzhou University Lanzhou 730000 China
| | - Leilei Ge
- Qingdao Vland Biotech Group Co., Ltd 266102 China
| | - Peng Wang
- College of Food Science and Engineering, Ocean University of China Qingdao 266071 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
| | - Ximing Xu
- Key Laboratory of Marine Drugs of Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Pilot National Laboratory for Marine Science and Technology Qingdao, Center for Innovation Marine Drug Screening & Evaluation Qingdao 266071 China
- Marine Drug Screening and Evaluation Platform (QNLM), Ocean University of China Qingdao 266071 China
| |
Collapse
|
35
|
Lin C, Zheng Q, Li Y, Wu T, Luo J, Jiang Y, Huang Q, Yan C, Zhang L, Zhang W, Liao H, Yang Y, Pang J. Assessment of the influence on left ventricle by potassium oxonate and hypoxanthine-induced chronic hyperuricemia. Exp Biol Med (Maywood) 2023; 248:165-174. [PMID: 36112877 PMCID: PMC10041052 DOI: 10.1177/15353702221120113] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cellular cytoplasmic xanthine oxidase (XO)-mediated uric acid synthesis and extracellular excess uric acid exposure are both causes of cardiomyocytic injury under the condition of hyperuricemia (HUA). Potassium oxonate suppresses uric acid degradation to increase extracellular concentration, while hypoxanthine is the catalytic substrate of XO. We aimed to observe cardiac damage in a chronic HUA mouse model induced by potassium oxonate and hypoxanthine. The mouse model was established by the co-administration of potassium oxonate and hypoxanthine for eight weeks. Then, left ventricular parameters were examined by echocardiographic evaluation, and the heart tissues were harvested for further histopathological analysis. The results showed that plasma uric acid was persistently elevated in the model mice, which demonstrated the stable establishment of chronic HUA. The left ventricular anterior wall was significantly thickened in the model group compared with the blank control group. After the end of modeling, the left ventricular anterior wall thickness of the hyperuricemic mice increased compared with that of blank group. The histological analysis showed and myocardial structure disorganization in the model group compared with the blank control. The above cardiac impairment changes could be attenuated by allopurinol pretreatment. This study systematically assessed cardiac damage in a chronic HUA mouse model. In addition, it provides useful information for future HUA-associated heart injury mechanism investigation and therapeutic treatment evaluation.
Collapse
Affiliation(s)
- Cuiting Lin
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Qiang Zheng
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yongmei Li
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Ting Wu
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jian Luo
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yanqing Jiang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Qinghua Huang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Caixin Yan
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Leqi Zhang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Wei Zhang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Hui Liao
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yang Yang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan 528000, Guangdong, China
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| |
Collapse
|
36
|
Zhong L, Lin Y, Gong S, Wu X, Liu Y, Chen J, Li Y, Yan F, Su Z, Xie Q. Oxyberberrubine, a novel liver microsomes-mediated secondary metabolite of berberine, alleviates hyperuricemic nephropathy in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154521. [PMID: 36334387 DOI: 10.1016/j.phymed.2022.154521] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/01/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Berberrubine (BRB), one of the major metabolites of berberine (BBR), exerts an anti-hyperuricemic effect even superior to BBR. Liver is an important location for drug transformation. Nevertheless, there are few studies on the bioactivities and metabolites of BRB. PURPOSE We investigated whether oxyberberrubine (OBR), a liver metabolite of BRB, exerted urate-lowering and reno-protective effects in hyperuricemic mice. METHODS Liver microsomes were used to incubate BRB for studying its biotransformation. We isolated and identified its new metabolite OBR, and investigated its anti-hyperuricemic and reno-protective effects. In this work, the hyperuricemic mice model was established by receiving potassium oxonate (PO) and hypoxanthine (HX) for 7 consecutive days. 1 h after modeling, different dosages of OBR (5, 10 and 20 mg/kg), BRB (20 mg/kg) or febuxostat (Fex, 5 mg/kg) were given mice by gavage. RESULTS Results showed that OBR possessed potent anti-hyperuricemic and reno-protective effects in hyperuricemic mice. Serum uric acid (UA) level was lowered, and the activities of xanthine oxidase (XOD) as well as adenosine deaminase (ADA) in the liver were suppressed after treatment with OBR. Hepatic expressions of XOD were remarkably decreased at mRNA and protein levels by OBR treatment. In addition, OBR prominently alleviated renal injury, embodied in markedly reduced serum creatinine and blood urea nitrogen (BUN) levels, decreased inflammatory mediators (TNF-α, IL-1β, IL-6 and IL-18) levels, mRNA expression of CYP27B1 and repairment of renal tissues damage. Besides, OBR down-regulated renal expression of urate transporter 1 (URAT1), glucose transporter 9 (GLUT9), NOD-like receptor 3 (NLRP3), apoptosis-associated speck-like protein containing CARD (ASC), and caspase-1 at mRNA and protein levels. CONCLUSIONS In short, our study indicated that OBR possessed superior anti-hyperuricemic and reno-protective effects, at least in part, through the inhibition of XOD, URAT1, GLUT9 and NLRP3 inflammasome signaling pathway in the kidney.
Collapse
Affiliation(s)
- Linjiang Zhong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Yinsi Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Shiting Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Xiaoyan Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Fan Yan
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Qingfeng Xie
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China; Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, PR China.
| |
Collapse
|
37
|
Zhang P, Sun H, Cheng X, Li Y, Zhao Y, Mei W, Wei X, Zhou H, Du Y, Zeng C. Dietary intake of fructose increases purine de novo synthesis: A crucial mechanism for hyperuricemia. Front Nutr 2022; 9:1045805. [PMID: 36601078 PMCID: PMC9807165 DOI: 10.3389/fnut.2022.1045805] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022] Open
Abstract
Background Fructose consumption is a potential risk factor for hyperuricemia because uric acid (UA) is a byproduct of fructose metabolism caused by the rapid consumption of adenosine triphosphate and accumulation of adenosine monophosphate (AMP) and other purine nucleotides. Additionally, a clinical experiment with four gout patients demonstrated that intravenous infusion of fructose increased the purine de novo synthesis rate, which implied fructose-induced hyperuricemia might be related to purine nucleotide synthesis. Moreover, the mechanistic (mammalian) target of rapamycin (mTOR) is a key protein both involved in fructose metabolism and purine de novo synthesis. The present study was conducted to elucidate how fructose influences mTOR and purine de novo synthesis in a hepatic cell line and livers of mice. Materials and methods RNA-sequencing in NCTC 1469 cells treated with 0- and 25-mM fructose for 24 h and metabolomics analysis on the livers of mice fed with 0- and 30-g/kg fructose for 2 weeks were assessed. Gene and protein expression of phosphoribosyl pyrophosphate synthase (PRPSAP1), Glutamine PRPP aminotransferase (PPAT), adenyl succinate lyase (ADSL), adenyl succinate synthetase isozyme-1 (Adss1), inosine-5'-monophosphate dehydrogenase (IMPDH), and guanine monophosphate synthetase (GMPS) was measured. The location of PRPSAP1 and PPAT in the liver was assessed by an immunofluorescence assay. Results Metabolite profiling showed that the level of AMP, adenine, adenosine, hypoxanthine, and guanine was increased significantly. RNA-sequencing showed that gene expression of phosphoribosyl pyrophosphate synthase (PRPS2), phosphoribosyl glycinamide formyl transferase (GART), AICAR transformylase (ATIC), ADSL, Adss1, and IMPDH were raised, and gene expression of adenosine monophosphate deaminase 3 (AMPD3), adenosine deaminase (ADA), 5',3'-nucleotidase, cytosolic (NT5C), and xanthine oxidoreductase (XOR) was also increased significantly. Fructose increased the gene expression, protein expression, and fluorescence intensity of PRPSAP1 and PPAT in mice livers by increasing mTOR expression. Fructose increased the expression and activity of XOR, decreased the expression of uricase, and increased the serum level of UA. Conclusion This study demonstrated that the increased purine de novo synthesis may be a crucial mechanism for fructose-induced hyperuricemia.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China,Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Huimin Sun
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Xinyu Cheng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Yajing Li
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Yanli Zhao
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Wuxuan Mei
- Clinical Medical College, Hubei University of Science and Technology, Xianning, China
| | - Xing Wei
- Department of Nephrotic Rheumatism, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, China
| | - Hairong Zhou
- Department of General Practice, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, China
| | - Yunbo Du
- Department of Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Changchun Zeng
- Department of General Practice, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, China,*Correspondence: Changchun Zeng,
| |
Collapse
|
38
|
Zhang J, Dong Y, Gao S, Zhang X, Liao H, Shi X, Zhang Z, Zhao T, Liang R, Qi D, Wu T, Pang J, Liu X, Zhan P. Design, synthesis and activity evaluation of novel lesinurad analogues containing thienopyrimidinone or pyridine substructure as human urate transporter 1 inhibitors. Eur J Med Chem 2022; 244:114816. [PMID: 36219903 DOI: 10.1016/j.ejmech.2022.114816] [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: 08/16/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022]
Abstract
Urate Transporter 1 (URAT1) plays a crucial role in uric acid transport, making it an attractive target for the treatment of gout and hyperuricemia. As a representative URAT1 inhibitor, Lesinurad treat gout by promoting the uric acid excretion. However, its lower in vitro and in vivo activity should be highly attracted attention. Herein, the bioisosterism, molecular hybridization and scaffold hopping strategies were exploited to modify all the structural components of Lesinurad and finally thirty novel compounds bearing thienopyrimidinone or pyridine core were obtained. Most of the compounds displayed certain URAT1 inhibitory activity in vitro. Among them, thienopyrimidinones 6 (IC50 = 7.68 μM), 10 (IC50 = 7.56 μM), 14 (IC50 = 7.31 μM) and 15 (IC50 = 7.90 μM) showed slightly better potency than positive control Lesinurad (IC50 = 9.38 μM). Notably, 10 also displayed inhibitory activity (IC50 = 55.96 μM) against GLUT9. Additionally, in vivo serum uric acid (SUA)-lowering experiments were performed on some representative compounds and it was revealed that all the selected compounds could decrease the SUA level in mice, of which the decrease rate of SUA was 73.29% for the most promising compound 10, significantly greater than that of Lesinurad (26.89%). Meanwhile, the preliminary SARs based on the URAT1 inhibitory activity were discussed in detail, which pointed out the direction for further structural optimization. Overall, the thienopyrimidinone and pyridine are prospective skeletons for the developing novel URAT1 inhibitors with considerable potential for optimization.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Yue Dong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Shenghua Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Xujie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Hui Liao
- School of Pharmaceutical Sciences, Southern Medical University, 1838 North Guangzhou Ave, 510515, Guangzhou, PR China
| | - Xiaoyu Shi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Zhijiao Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Tong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Ruipeng Liang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Danhui Qi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Ting Wu
- School of Pharmaceutical Sciences, Southern Medical University, 1838 North Guangzhou Ave, 510515, Guangzhou, PR China.
| | - Jianxin Pang
- School of Pharmaceutical Sciences, Southern Medical University, 1838 North Guangzhou Ave, 510515, Guangzhou, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| |
Collapse
|
39
|
Shi X, Zhao T, da Silva-Júnior EF, Zhang J, Xu S, Gao S, Liu X, Zhan P. Novel urate transporter 1 (URAT1) inhibitors: a review of recent patent literature (2020-present). Expert Opin Ther Pat 2022; 32:1175-1184. [PMID: 36625031 DOI: 10.1080/13543776.2022.2165911] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The urate transporter 1 (URAT1) is a membrane transporter located in the apical membrane of human renal proximal tubule epithelial cells, which mediates most of the reabsorption of urate. Hyperuricemia (HUA) is a common disease caused by metabolic disorders, which has been considered as the key factor of gout. Approximately 90% of patients suffer from hyperuricemia due to insufficient or poor uric acid excretion. Therefore, the drug design of URAT1 inhibitors targeting improve the renal urate excretion by reducing the reabsorption of urate anions represent a hot topic in searching for anti-gout drugs currently. AREAS COVERED In this review, we summarize URAT1 inhibitors patents reported since 2020 to present through the public database at https://worldwide.espacenet.com and some medicinal chemistry strategies employed to develop novel drug candidates. EXPERT OPINION Ligand-based drug design (LBDD) strategies have been frequently used developing new URAT1 inhibitors. Meanwhile, the discovery of dual drugs targeting both inhibition of xanthine oxidase (XOD) and URAT1 may be an emerging horizon for designing novel uric acid-lowering candidates in future. Furthermore, advanced techniques in the field of molecular biology and computer science can increase the chances to discover and/or optimize URAT1 inhibitors, contributing to the development of novel drug candidates.
Collapse
Affiliation(s)
- Xiaoyu Shi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Shandong, PR China
| | - Tong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Shandong, PR China
| | | | - Jian Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Shandong, PR China
| | - Shujing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Shandong, PR China
| | - Shenghua Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Shandong, PR China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Shandong, PR China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Shandong, PR China
| |
Collapse
|
40
|
Uricase-Deficient Larval Zebrafish with Elevated Urate Levels Demonstrate Suppressed Acute Inflammatory Response to Monosodium Urate Crystals and Prolonged Crystal Persistence. Genes (Basel) 2022; 13:genes13122179. [PMID: 36553446 PMCID: PMC9777727 DOI: 10.3390/genes13122179] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Gout is caused by elevated serum urate leading to the deposition of monosodium urate (MSU) crystals that can trigger episodes of acute inflammation. Humans are sensitive to developing gout because they lack a functional urate-metabolizing enzyme called uricase/urate oxidase (encoded by the UOX gene). A hallmark of long-standing disease is tophaceous gout, characterized by the formation of tissue-damaging granuloma-like structures ('tophi') composed of densely packed MSU crystals and immune cells. Little is known about how tophi form, largely due to the lack of suitable animal models in which the host response to MSU crystals can be studied in vivo long-term. We have previously described a larval zebrafish model of acute gouty inflammation where the host response to microinjected MSU crystals can be live imaged within an intact animal. Although useful for modeling acute inflammation, crystals are rapidly cleared following a robust innate immune response, precluding analysis at later stages. Here we describe a zebrafish uox null mutant that possesses elevated urate levels at larval stages. Uricase-deficient 'hyperuricemic' larvae exhibit a suppressed acute inflammatory response to MSU crystals and prolonged in vivo crystal persistence. Imaging of crystals at later stages reveals that they form granuloma-like structures dominated by macrophages. We believe that uox-/- larvae will provide a useful tool to explore the transition from acute gouty inflammation to tophus formation, one of the remaining mysteries of gout pathogenesis.
Collapse
|
41
|
Allegrini S, Garcia-Gil M, Pesi R, Camici M, Tozzi MG. The Good, the Bad and the New about Uric Acid in Cancer. Cancers (Basel) 2022; 14:cancers14194959. [PMID: 36230882 PMCID: PMC9561999 DOI: 10.3390/cancers14194959] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The concentration of uric acid in blood is sex-, age- and diet-dependent and is maintained close to its maximal solubility, indicating that it plays some important role. Indeed, it has been demonstrated that, at physiological concentrations, uric acid is a powerful antioxidant and is a scavenger of singlet oxygen and radicals. At high intracellular concentration, uric acid has been demonstrated to act as a pro-oxidant molecule. Recently, uric acid has been reported to affect the properties of several proteins involved in metabolic regulation and signaling, and the relationship between uric acid and cancer has been extensively investigated. In this review, we present the most recent results on the positive and negative effects played by uric acid in cancer and some new findings and hypotheses about the implication of this metabolite in the pathogenesis of several diseases such as metabolic syndrome, diabetes, and inflammation, thus favoring the development of cancer. Abstract Uric acid is the final product of purine catabolism in man and apes. The serum concentration of uric acid is sex-, age- and diet-dependent and is maintained close to its maximal solubility, indicating that it plays some important role. Indeed, it has been demonstrated that, at physiological concentrations, uric acid is a powerful antioxidant, while at high intracellular concentrations, it is a pro-oxidant molecule. In this review, we describe the possible causes of uric acid accumulation or depletion and some of the metabolic and regulatory pathways it may impact. Particular attention has been given to fructose, which, because of the complex correlation between carbohydrate and nucleotide metabolism, causes uric acid accumulation. We also present recent results on the positive and negative effects played by uric acid in cancer and some new findings and hypotheses about the implication of this metabolite in a variety of signaling pathways, which can play a role in the pathogenesis of diseases such as metabolic syndrome, diabetes, and inflammation, thus favoring the development of cancer. The loss of uricase in Homo sapiens and great apes, although exposing these species to the potentially adverse effects of uric acid, appears to be associated with evolutionary advantages.
Collapse
Affiliation(s)
- Simone Allegrini
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, Università di Pisa, 56126 Pisa, Italy
- CISUP, Centro per L’Integrazione della Strumentazione dell’Università di Pisa, 56127 Pisa, Italy
- Correspondence:
| | - Mercedes Garcia-Gil
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, Università di Pisa, 56126 Pisa, Italy
- CISUP, Centro per L’Integrazione della Strumentazione dell’Università di Pisa, 56127 Pisa, Italy
- Unità di Fisiologia Generale, Dipartimento di Biologia, Università di Pisa, Via San Zeno 31, 56127 Pisa, Italy
| | - Rossana Pesi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| | - Marcella Camici
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| | - Maria Grazia Tozzi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| |
Collapse
|
42
|
Wang S, Zhang L, Hao D, Wang L, Liu J, Niu Q, Mi L, Peng X, Gao J. Research progress of risk factors and early diagnostic biomarkers of gout-induced renal injury. Front Immunol 2022; 13:908517. [PMID: 36203589 PMCID: PMC9530830 DOI: 10.3389/fimmu.2022.908517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Gout renal injury has an insidious onset, no obvious symptoms, and laboratory abnormalities in the early stages of the disease. The injury is not easily detected, and in many cases, the patients have entered the renal failure stage at the time of diagnosis. Therefore, the detection of gout renal injury–related risk factors and early diagnostic biomarkers of gout renal injury is essential for the prevention and early diagnosis of the disease. This article reviews the research progress in risk factors and early diagnostic biomarkers of gout renal injury.
Collapse
Affiliation(s)
- Sheng Wang
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, China
| | - Liyun Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, China
| | - Dongsheng Hao
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, China
| | - Lei Wang
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, China
| | - Jiaxi Liu
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, China
| | - Qing Niu
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Liangyu Mi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xinyue Peng
- Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, China
- *Correspondence: Jinfang Gao,
| |
Collapse
|
43
|
lncRNA TUG1 regulates hyperuricemia-induced renal fibrosis in a rat model. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1365-1375. [PMID: 36148952 PMCID: PMC9828301 DOI: 10.3724/abbs.2022128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Renal fibrosis is most common among chronic kidney diseases. Molecular studies have shown that long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) participate in renal fibrosis, while the roles of lncRNA taurine upregulated gene 1 (TUG1) and miR-140-3p in hyperuricemia-induced renal fibrosis remain less investigated. In this study, a rat hyperuricemia model is constructed by oral administration of adenine. TUG1, miR-140-3p, and cathepsin D (CtsD) expression levels in rat models are measured. After altering TUG1, miR-140-3p, or CtsD expression in modelled rats, biochemical indices, including uric acid (UA), serum creatine (SCr), blood urea nitrogen (BUN), and 24-h urine protein are detected, pathological changes in the renal tissues, and renal fibrosis are examined. In renal tissues from hyperuricemic rats, TUG1 and CtsD are upregulated, while miR-140-3p is downregulated. Inhibiting TUG1 or CtsD or upregulating miR-140-3p relieves renal fibrosis in hyperuricemic rats. Downregulated miR-140-3p reverses the therapeutic effect of TUG1 reduction, while overexpression of CtsD abolishes the role of miR-140-3p upregulation in renal fibrosis. Collectively, this study highlights that TUG1 inhibition upregulates miR-140-3p to ameliorate renal fibrosis in hyperuricemic rats by inhibiting CtsD.
Collapse
|
44
|
Xu Z, Sha W, Hou C, Amakye WK, Yao M, Ren J. Comparison of 3 hyperuricemia mouse models and evaluation of food-derived anti-hyperuricemia compound with spontaneous hyperuricemia mouse model. Biochem Biophys Res Commun 2022; 630:41-49. [PMID: 36137324 DOI: 10.1016/j.bbrc.2022.09.043] [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: 08/31/2022] [Revised: 09/04/2022] [Accepted: 09/10/2022] [Indexed: 11/30/2022]
Abstract
Hyperuricemia animal models have long been used for evaluating food-derived anti-hyperuricemia compounds. Fructose and potassium oxonate are commonly used for developing hyperuricemia mouse model. Recent research also developed spontaneous hyperuricemia model by uricase knockout (Uox-/-). In this work, we evaluated 3 kinds of models with the same gene background to illustrate the differences between the treatments. Unlike the uric acid levels in potassium oxonate (224.79 ± 33.62 μmol/L) and Uox-/- groups (458.39 ± 38.29 μmol/L), fructose treatment did not lead to higher serum uric acid level (174.93 ± 30.46 μmol/L) comparing to the control group (153.53 ± 40.96 μmol/L). However, abnormal glycometabolism only developed in the fructose and the Uox-/- group. In addition, anemia, inflammasome and severe renal injury occurred in the Uox-/- group. The Uox-/- mice were then treated with puerarin and allopurinol, and found that puerarin could reduce serum uric acid and alleviated the serious renal damage associated with high uric acid. Thus, the Uox-/- mice could be a suitable model for screening and evaluating anti-hyperuricemia compounds.
Collapse
Affiliation(s)
- Zhenzhen Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China; China-Singapore International Joint Research Institute, Guangzhou Knowledge City, Huangpu District, Guangzhou, 510663, China
| | - Wanqian Sha
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Chuanli Hou
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - William Kwame Amakye
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Maojin Yao
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease, Guangzhou, 510182, China
| | - Jiaoyan Ren
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China; China-Singapore International Joint Research Institute, Guangzhou Knowledge City, Huangpu District, Guangzhou, 510663, China.
| |
Collapse
|
45
|
|
46
|
Zeng H, Tang C, Lin B, Yu M, Wang X, Wang J, Chen S, Yu C. The regulation effect of GLUT9/SLC2A9 on intrahepatic uric acid level and metabolic associated fatty liver disease. Hepatol Int 2022; 16:1064-1074. [PMID: 36006548 DOI: 10.1007/s12072-022-10371-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/22/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Metabolic associated fatty liver disease (MAFLD) is the most common chronic liver disease worldwide. The important role of urid acid (UA) in MAFLD has been widely investigated. Our previous studies unveiled the elevation of serum UA levels independently predicts an increased risk of incident MAFLD. However, the role of intrahepatic UA in MAFLD has not been investigated yet. Glucose transporter 9 (GLUT9) is a key transporter that mediates the uptake of UA in hepatocytes. METHODS In this study, we first explored the clinical association between GLUT9 polymorphism and MAFLD. Blood samples of 247 male Chinese (127 were MAFLD patients) were collected and tested for the blood UA levels and genotype of the single nucleotide polymorphism (SNP) of GLUT9 (rs1014290). Next, Glut9 hepatic-specific knockout mice (Glut9Hep-ko) were generated to investigate the role of hepatic GLUT9 in MAFLD in male mice. RESULTS We found that the GA/AA genotypes (rs1014290) were associated with elevated serum UA levels in MAFLD patients. Meanwhile, we found that Glut9Hep-ko mice displayed lower intrahepatic UA levels, down-regulated lipogenesis genes expressions, and attenuated MAFLD symptoms after 12 weeks of high-fat diet feeding, compared with Glut9Fl/Fl littermates. However, Glut9Hep-ko mice and wild-type littermates showed no significant difference on hepatic fatty acid oxidation or inflammation. CONCLUSIONS Our results suggested that GLUT9 polymorphism was significantly associated with MAFLD, and hepatic-specific knockout of Glut9 significantly decreased intrahepatic contents and ameliorated diet-induced MAFLD in mice.
Collapse
Affiliation(s)
- Hang Zeng
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| | - Chenxi Tang
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Bingru Lin
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Mengli Yu
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xinyu Wang
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jinghua Wang
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Shenghui Chen
- Department of Gastroenterology, School of Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University, Hangzhou, 310003, China
| | - Chaohui Yu
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
47
|
Qi X, Chen H, Guan K, Sun Y, Wang R, Li Q, Ma Y. Novel xanthine oxidase inhibitory peptides derived from whey protein: identification, in vitro inhibition mechanism and in vivo activity validation. Bioorg Chem 2022; 128:106097. [PMID: 35985156 DOI: 10.1016/j.bioorg.2022.106097] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 11/28/2022]
Abstract
As the development of hyperuricemia (HUA) and gout continues to accelerate worldwide, there is increasing interest in the use of xanthine oxidase (XO) inhibitors as therapeutic agents for the management of HUA and gout. In the present study, XO inhibitory peptides were identified from whey protein isolate (WPI) hydrolysates, and the underlying inhibitory mechanism and in vivo activities was investigated. WPI hydrolysates were isolated and purified, and two peptides (ALPM and LWM) with lower binding energy were screened by molecular docking. The result showed that these two peptides interacted with residues around the active site of XO through hydrogen bond and hydrophobic interaction. The IC50 values of ALPM and LWM were 7.23 ± 0.22 and 5.01 ± 0.31 mM, respectively. According to the Lineweaver-Burk curve, the inhibition types of ALPM and LWM were non-competitive inhibition. Circular dichroism (CD) spectra indicated ALPM and LWM could change the secondary structure of XO. Molecular dynamics simulations revealed that XO-peptide complexes were more stable and compact than XO. Moreover, animal studies have shown that ALPM and LWM have anti-hyperuricemia effects in vivo. This study suggested that ALPM and LWM can be considered as natural XO inhibitors for the treatment of HUA.
Collapse
Affiliation(s)
- Xiaofen Qi
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Haoran Chen
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Kaifang Guan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Yue Sun
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Rongchun Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Qiming Li
- New Hope Dairy Co, Ltd, Chengdu 610063, Sichuan, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
48
|
Cao J, Bu Y, Hao H, Liu Q, Wang T, Liu Y, Yi H. Effect and Potential Mechanism of Lactobacillus plantarum Q7 on Hyperuricemia in vitro and in vivo. Front Nutr 2022; 9:954545. [PMID: 35873427 PMCID: PMC9298507 DOI: 10.3389/fnut.2022.954545] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/13/2022] [Indexed: 12/14/2022] Open
Abstract
Hyperuricemia (HUA) is a disorder of purine metabolism resulting in abnormally elevated serum uric acid (UA) concentration. It is believed that there is an association between gut microbiota and HUA, and probiotics have the potential palliative effect. However, the underlying mechanism of probiotics in ameliorating HUA remains unclear. The purpose of this study was to investigate the effect and mechanism of Lactobacillus plantarum Q7 on HUA in Balb/c mice. The results showed that L. plantarum Q7 had an excellent capability to affect UA metabolism, which could degrade nucleotides by 99.97%, nucleosides by 99.15%, purine by 87.35%, and UA by 81.30%. It was observed that L. plantarum Q7 could downregulate serum UA, blood urea nitrogen (BUN), creatinine (Cr), and xanthine oxidase (XOD) by 47.24%, 14.59%, 54.59%, and 40.80%, respectively. Oral administration of L. plantarum Q7 could restore the liver, kidney, and intestinal injury induced by HUA and the expression of metabolic enzymes and transporters to normal level. 16S rRNA sequencing analysis showed that L. plantarum Q7 treatment could restore the imbalance of species diversity, richness, and community evenness compared with the model group. The ratio of Bacteroidetes to Firmicutes was recovered nearly to the normal level by L. plantarum Q7 intervention. The dominant microorganisms of L. plantarum Q7 group contained more anti-inflammatory bacteria than those of the model group. These findings indicated that L. plantarum Q7 might regulate UA metabolism and repair the liver and kidney injury by reshaping the gut microbiota and could be used as a potential probiotic strain to ameliorate HUA.
Collapse
Affiliation(s)
- Jiayuan Cao
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yushan Bu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Haining Hao
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Qiqi Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Ting Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yisuo Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
49
|
Zhang W, Cui Y, Zhang J. Multi metabolomics-based analysis of application of Astragalus membranaceus in the treatment of hyperuricemia. Front Pharmacol 2022; 13:948939. [PMID: 35935868 PMCID: PMC9355468 DOI: 10.3389/fphar.2022.948939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/29/2022] [Indexed: 11/15/2022] Open
Abstract
Hyperuricemia (HUA) is a common metabolic disease that is an independent risk factor for comorbidities such as hypertension, chronic kidney disease, and coronary artery disease. The prevalence of HUA has increased over the last several decades with improved living standards and increased lifespans. Metabolites are considered the most direct reflection of individual physiological and pathological conditions, and represent attractive candidates to provide deep insights into disease phenotypes. Metabolomics, a technique used to profile metabolites in biofluids and tissues, is a powerful tool for identification of novel biomarkers, and can be used to provide valuable insights into the etiopathogenesis of metabolic diseases and to evaluate the efficacy of drugs. In this study, multi metabolomics-based analysis of the blood, urine, and feces of rats with HUA showed that HUA significantly altered metabolite profiles. Astragalus membranaceus (AM) and benbromomalone significantly mitigated these changes in blood and feces, but not in urine. Some crucial metabolic pathways including lipid metabolism, lipid signaling, hormones synthesis, unsaturated fatty acid (UFAs) absorption, and tryptophan metabolism, were seriously disrupted in HUA rats. In addition, AM administration exerted better treatment effects on HUA than benbromomalone. Furthermore, additional supplementation with UFAs and tryptophan may also induce therapeutic effects against HUA.
Collapse
Affiliation(s)
- Wenwen Zhang
- The School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yifang Cui
- The School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiayu Zhang
- The School of Pharmacy, Binzhou Medical University, Yantai, China
- *Correspondence: Jiayu Zhang,
| |
Collapse
|
50
|
Geng F, Chen J, Tang S, Azzam E, Zhang J, Zhang S. Additional Evidence for Commonalities between COVID-19 and Radiation Injury: Novel Insight into COVID-19 Candidate Drugs. Radiat Res 2022; 198:306-317. [PMID: 35834824 DOI: 10.1667/rade-22-00058.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/14/2022] [Indexed: 11/03/2022]
Abstract
COVID-19 is a challenge to biosecurity and public health. The speed of vaccine development lags behind that of virus evolution and mutation. To date, no agent has been demonstrated to be fully effective against COVID-19. Therefore, it remains of great urgency to rapidly develop promising therapeutic and diagnostic candidates. Intriguingly, mounting evidence hints at parallel etiologies between SARS-CoV-2 infection and radiation injury. Herein, from the perspectives of immunogenic pathway activation and metabolic alterations, we provide novel evidence of commonalities between these two pathological conditions based on the most recent findings. Since numerous agents have been developed to prevent or reverse radiation injury in the past 70 years to ensure nuclear safety, we also advocate investigating the promising function of radioprotectors and radiomitigators against COVID-19 in clinical settings.
Collapse
Affiliation(s)
- Fenghao Geng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.,West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Jianhui Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Shaokai Tang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Edouard Azzam
- Radiobiology and Health, Isotopes, Radiobiology & Environment Directorate (IRED), Canadian Nuclear Laboratories (CNL), Chalk River, ON K0J 1J0, Canada
| | - Jie Zhang
- Institute of Preventive Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Shuyu Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.,West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China.,NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang 621099, China
| |
Collapse
|