1
|
Nakayama S, Satoh M, Toyama M, Hashimoto H, Murakami T, Hirose T, Obara T, Mori T, Metoki H. Comparison of the incidence of proteinuria and changes in eGFR among febuxostat and topiroxostat users. Clin Exp Nephrol 2025:10.1007/s10157-025-02630-x. [PMID: 39881083 DOI: 10.1007/s10157-025-02630-x] [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: 06/13/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Febuxostat and topiroxostat are non-purine selective xanthine oxidoreductase inhibitors commonly used for hyperuricaemia treatment in Japan. However, comparative data on the effects of febuxostat and topiroxostat on renal function and proteinuria are limited. This study compared proteinuria incidence and changes in the estimated glomerular filtration rate (eGFR) among prevalent febuxostat and topiroxostat users. METHODS We conducted a retrospective cohort study using databases provided by DeSC Healthcare, Inc. (Tokyo, Japan). We identified 17,446 individuals (11.8% women; mean age 67.4 years) with eGFR ≥ 30 mL/min/1.73 m2 and no history of cardiovascular disease or proteinuria at baseline. Separate analyses were performed for individuals with eGFR < 60 mL/min/1.73 m2 and those with eGFR ≥ 60 mL/min/1.73 m2. The adjusted hazard ratio (HR) for proteinuria incidence in topiroxostat users compared with febuxostat users was assessed using the Cox model. Changes in eGFR were compared between the two groups using multiple regression analysis. RESULTS During the mean follow-up period of 1.79 years, 1,433 participants developed proteinuria. In non-diabetic individuals with eGFR ≥ 60 mL/min/1.73 m2, the adjusted HR for proteinuria incidence in topiroxostat users compared with febuxostat users was 0.60 (95% confidence interval, 0.40-0.91; p = 0.016). No significant differences were observed in eGFR changes between the two groups with eGFR < 60 and ≥ 60 mL/min/1.73 m2. CONCLUSION Topiroxostat prevalent users had a lower risk of proteinuria than febuxostat prevalent users in non-diabetic individuals with eGFR ≥ 60 mL/min/1.73 m2. Our findings suggest that topiroxostat might be more effective than febuxostat in preventing proteinuria in non-diabetic individuals with eGFR ≥ 60 mL/min/1.73 m2.
Collapse
Affiliation(s)
- Shingo Nakayama
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Michihiro Satoh
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.
- Department of Pharmacy, Tohoku Medical and Pharmaceutical University Hospital, Sendai, Japan.
| | - Maya Toyama
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Department of Nephrology, Self-Defense Forces Sendai Hospital, Sendai, Japan
| | - Hideaki Hashimoto
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Takahisa Murakami
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Division of Aging and Geriatric Dentistry, Department of Rehabilitation Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Takuo Hirose
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Obara
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Takefumi Mori
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Hirohito Metoki
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Tohoku Institute for Management of Blood Pressure, Sendai, Japan
| |
Collapse
|
2
|
Choi C, Kim MG, Kim JH. Reno-protective effects of xanthine oxidase inhibitors in patients with type 2 diabetes and chronic kidney disease: a systematic review and meta-analysis. J Nephrol 2025:10.1007/s40620-024-02199-w. [PMID: 39865217 DOI: 10.1007/s40620-024-02199-w] [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: 09/27/2024] [Accepted: 12/15/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The effect of lowering uric acid levels on renal function in patients with diabetic kidney disease remains unclear. Previous randomized controlled trials (RCTs) have reported conflicting results regarding the effects of xanthine oxidase inhibitors on renal function. This study aimed to examine the renoprotective effects of xanthine oxidase inhibitors (febuxostat and topiroxostat) in patients with diabetic kidney disease. METHODS Relevant RCTs were searched using PubMed, Embase, and Cochrane Central databases. Ultimately, five RCTs were included in the meta-analysis. The assessed renal endpoints included changes in the estimated glomerular filtration rate (eGFR) and urine albumin-to-creatinine ratio. The meta-analysis was conducted using Review Manager version 5.4. Standardized mean differences (SMDs) and 95% confidence intervals (CIs) were calculated for changes in renal endpoints between the groups after the study period. A subgroup analysis was conducted based on the type of intervention, results of the risk of bias assessment, and baseline renal function. RESULTS Although the use of febuxostat or topiroxostat did not induce a significant change in eGFR compared with the placebo, it showed a tendency to delay renal function decline (SMD = 0.32, 95% CI = [- 0.00; 0.64]). There was no significant difference in albuminuria between the two groups (SMD = 0.26, 95% CI = [- 0.10; 0.62]). CONCLUSIONS This study suggests the potential of febuxostat or topiroxostat to delay renal function decline in patients with diabetes and underlying renal impairment, that needs to be confirmed in further studies. TRIAL REGISTRATION INPLASY registration number 202450024.
Collapse
Affiliation(s)
- Chiwon Choi
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju, 54907, Republic of Korea
| | - Myeong Gyu Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
- College of Pharmacy, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jae Hyun Kim
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju, 54907, Republic of Korea.
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, 54907, Republic of Korea.
| |
Collapse
|
3
|
Hagiwara M, Ishiyama S, Nakamura T, Mochizuki K. Topiroxostat improves glomerulosclerosis in type 2 diabetic Nagoya Shibata Yasuda mice with early diabetic kidney disease. Eur J Pharmacol 2024; 982:176915. [PMID: 39154822 DOI: 10.1016/j.ejphar.2024.176915] [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: 02/24/2024] [Revised: 07/12/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Reactive oxygen species production might be prevented by xanthine oxidoreductase (XOR) inhibitors, which can cause glomerulosclerosis. We aimed to investigate whether topiroxostat, an XOR inhibitor, prevents diabetic kidney disease development in mice. Six-week-old control Institute of Cancer Research (ICR) mice and type 2 diabetic Nagoya Shibata Yasuda (NSY) mice were divided into the ICR group (ICR mice which received a lard-containing high-fat diet [HFD] based on the AIN-93G diet), NSY control group (NSY mice which received the same aforementioned diet), and NSY + topiroxostat group (NSY mice which received the same aforementioned diet with addition of 0.0012% topiroxostat). After 20 weeks, plasma biomarkers, XOR activity and oxidative stress levels, which were assessed using malondialdehyde (MDA), were measured through enzyme-linked immunosorbent assay or enzymatic methods. Renal pathology was evaluated using periodic acid-Schiff staining. Redox gene and protein expression were determined using RT-qPCR and western blotting, respectively. Plasma XOR activity was lower in NSY mice treated with topiroxostat than those without. Plasma cystatin C and creatinine levels did not differ between the ICR and NSY control groups or between the NSY control and NSY + topiroxostat groups. The NSY + topiroxostat group showed a smaller mesangial area than the NSY control group. The mRNA expression of Sod3, Prdx1, Gpx2, and Gpx3 was higher in the NSY + topiroxostat group than in the NSY control group. Renal MDA levels were lower in the NSY + topiroxostat group than in the NSY control group. Topiroxostat can reduce glomerulosclerosis, and the reduction is associated with renal oxidative markers.
Collapse
Affiliation(s)
- Mai Hagiwara
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Yamanashi, Japan.
| | - Shiori Ishiyama
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Yamanashi, Japan; Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, Japan.
| | - Takashi Nakamura
- Pharmacological Study Group Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho, Mie, Japan.
| | - Kazuki Mochizuki
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Yamanashi, Japan; Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, Japan.
| |
Collapse
|
4
|
Korsmo HW, Ekperikpe US, Daehn IS. Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:712. [PMID: 38929151 PMCID: PMC11200862 DOI: 10.3390/antiox13060712] [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: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Xanthine Oxidoreductase (XOR) is a ubiquitous, essential enzyme responsible for the terminal steps of purine catabolism, ultimately producing uric acid that is eliminated by the kidneys. XOR is also a physiological source of superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of various physiological pathways, as well as contribute to the development and the progression of chronic conditions including kidney diseases, which are increasing in prevalence worldwide. XOR activity can promote oxidative distress, endothelial dysfunction, and inflammation through the biological effects of reactive oxygen species; nitric oxide and uric acid are the major products of XOR activity. However, the complex relationship of these reactions in disease settings has long been debated, and the environmental influences and genetics remain largely unknown. In this review, we give an overview of the biochemistry, biology, environmental, and current clinical impact of XOR in the kidney. Finally, we highlight recent genetic studies linking XOR and risk for kidney disease, igniting enthusiasm for future biomarker development and novel therapeutic approaches targeting XOR.
Collapse
Affiliation(s)
| | | | - Ilse S. Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1243, New York, NY 10029, USA
| |
Collapse
|
5
|
Yang YS, Wang B, Liu J, Li Q, Jiao QC, Qin P. Discovery of coumaric acid derivatives hinted by coastal marine source to seek for uric acid lowering agents. J Enzyme Inhib Med Chem 2023; 38:2163241. [PMID: 36629443 PMCID: PMC9848256 DOI: 10.1080/14756366.2022.2163241] [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] [Indexed: 01/12/2023] Open
Abstract
In this work, a series of novel compounds Spartinin C1-C24 were screened, synthesised and evaluated for inhibiting xanthine oxidase thus lowering serum uric acid level. The backbones were derived from the components of coastal marine source Spartina alterniflora and marketed drugs. The top hits Spartinin C10 & C22 suggested high inhibition percentages (78.54 and 93.74) at 10 μM dosage, which were higher than the positive control Allopurinol. They were low cytotoxic onto human normal hepatocyte cells. Treatment with Spartinin C10 could lower the serum uric acid level to 440.0 μM in the hyperuricemic model mice (723.0 μM), comparable with Allopurinol (325.8 μM). Spartinin C10 was more appreciated than Allopurinol on other serum indexes. The preliminary pharmacokinetics evaluation indicated that the rapid absorption, metabolism and elimination of Spartinin C10 should be further improved. The discovery of pharmaceutical molecules from coastal marine source here might inspire the inter-disciplinary investigations on public health.
Collapse
Affiliation(s)
- Yu-Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China,Research and Development Center, Nanjing Shibeitai Biotechnology Co., Ltd., Nanjing, China,Yu-Shun Yang State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Bin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Junzhong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qin Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qin-Cai Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China,CONTACT Qin-Cai Jiao
| | - Pei Qin
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China,Research and Development Center, Nanjing Shibeitai Biotechnology Co., Ltd., Nanjing, China,Pei Qin
| |
Collapse
|
6
|
Wang Y, Zhou L, Chen M, Liu Y, Yang Y, Lu T, Ban F, Hu X, Qian Z, Hong P, Zhang Y. Mining Xanthine Oxidase Inhibitors from an Edible Seaweed Pterocladiella capillacea by Using In Vitro Bioassays, Affinity Ultrafiltration LC-MS/MS, Metabolomics Tools, and In Silico Prediction. Mar Drugs 2023; 21:502. [PMID: 37888437 PMCID: PMC10608504 DOI: 10.3390/md21100502] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023] Open
Abstract
The prevalence of gout and the adverse effects of current synthetic anti-gout drugs call for new natural and effective xanthine oxidase (XOD) inhibitors to target this disease. Based on our previous finding that an edible seaweed Pterocladiella capillacea extract inhibits XOD, XOD-inhibitory and anti-inflammatory activities were used to evaluate the anti-gout potential of different P. capillacea extract fractions. Through affinity ultrafiltration coupled with liquid chromatography tandem mass spectrometry (LC-MS/MS), feature-based molecular networking (FBMN), and database mining of multiple natural products, the extract's bioactive components were traced and annotated. Through molecular docking and ADMET analysis, the possibility and drug-likeness of the annotated XOD inhibitors were predicted. The results showed that fractions F4, F6, F4-2, and F4-3 exhibited strong XOD inhibition activity, among which F4-3 reached an inhibition ratio of 77.96% ± 4.91% to XOD at a concentration of 0.14 mg/mL. In addition, the P. capillacea extract and fractions also displayed anti-inflammatory activity. Affinity ultrafiltration LC-MS/MS analysis and molecular networking showed that out of the 20 annotated compounds, 8 compounds have been previously directly or indirectly reported from seaweeds, and 4 compounds have been reported to exhibit anti-gout activity. Molecular docking and ADMET showed that six seaweed-derived compounds can dock with the XOD activity pocket and follow the Lipinski drug-like rule. These results support the value of further investigating P. capillacea as part of the development of anti-gout drugs or related functional foods.
Collapse
Affiliation(s)
- Yawen Wang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Longjian Zhou
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Minqi Chen
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Yayue Liu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yu Yang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Tiantian Lu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Fangfang Ban
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Xueqiong Hu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Zhongji Qian
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
| | - Pengzhi Hong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yi Zhang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
7
|
Sato K, Naganuma A, Nagashima T, Arai Y, Mikami Y, Nakajima Y, Kanayama Y, Murakami T, Uehara S, Uehara D, Yamazaki Y, Murase T, Nakamura T, Uraoka T. A Newly Developed Method-Based Xanthine Oxidoreductase Activities in Various Human Liver Diseases. Biomedicines 2023; 11:biomedicines11051445. [PMID: 37239117 PMCID: PMC10216503 DOI: 10.3390/biomedicines11051445] [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: 03/11/2023] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Studies evaluating xanthine oxidoreductase (XOR) activities in comprehensive liver diseases are scarce, and different etiologies have previously been combined in groups for comparison. To accurately evaluate XOR activities in liver diseases, the plasma XOR activities in etiology-based comprehensive liver diseases were measured using a novel, sensitive, and accurate assay that is a combination of liquid chromatography and triple quadrupole mass spectrometry to detect [13C2, 15N2]uric acid using [13C2, 15N2]xanthine as a substrate. We also mainly evaluated the association between the plasma XOR activities and parameters of liver tests, purine metabolism-associated markers, oxidative stress markers, and an inflammation marker. In total, 329 patients and 32 controls were enrolled in our study. Plasma XOR activities were generally increased in liver diseases, especially in the active phase, such as in patients with hepatitis C virus RNA positivity, those with abnormal alanine transaminase (ALT) levels in autoimmune liver diseases, and uncured hepatocellular carcinoma patients. Plasma XOR activities were numerically highest in patients with acute hepatitis B. Plasma XOR activities were closely correlated with parameters of liver tests, especially serum ALT levels, regardless of etiology and plasma xanthine levels. Our results indicated that plasma XOR activity might reflect the active phase in various liver diseases.
Collapse
Affiliation(s)
- Ken Sato
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
- Department of Hepatology, Heisei Hidaka Clinic, Takasaki 371-0001, Japan
- Department of Healthcare Informatics, Takasaki University of Health and Welfare, Takasaki 370-0033, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki 370-0829, Japan
| | - Tamon Nagashima
- Department of Gastroenterology, National Hospital Organization Shibukawa Medical Center, Shibukawa 377-0204, Japan
| | - Yosuke Arai
- Department of Gastroenterology, National Hospital Organization Shibukawa Medical Center, Shibukawa 377-0204, Japan
| | - Yuka Mikami
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yuka Nakajima
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yuki Kanayama
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Tatsuma Murakami
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki 370-0829, Japan
| | - Sanae Uehara
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki 370-0829, Japan
| | - Daisuke Uehara
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yuichi Yamazaki
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Takayo Murase
- Mie Research Park, Sanwa Kagaku Kenkyusho, Inabe 511-0406, Japan
| | - Takashi Nakamura
- Mie Research Park, Sanwa Kagaku Kenkyusho, Inabe 511-0406, Japan
| | - Toshio Uraoka
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| |
Collapse
|
8
|
Maghsoud Y, Dong C, Cisneros GA. Computational Characterization of the Inhibition Mechanism of Xanthine Oxidoreductase by Topiroxostat. ACS Catal 2023; 13:6023-6043. [PMID: 37547543 PMCID: PMC10399974 DOI: 10.1021/acscatal.3c01245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Xanthine oxidase (XO) is a member of the molybdopterin-containing enzyme family. It interconverts xanthine to uric acid as the last step of purine catabolism in the human body. The high uric acid concentration in the blood directly leads to human diseases like gout and hyperuricemia. Therefore, drugs that inhibit the biosynthesis of uric acid by human XO have been clinically used for many years to decrease the concentration of uric acid in the blood. In this study, the inhibition mechanism of XO and a new promising drug, topiroxostat (code: FYX-051), is investigated by employing molecular dynamics (MD) and quantum mechanics/molecular mechanics (QM/MM) calculations. This drug has been reported to act as both a noncovalent and covalent inhibitor and undergoes a stepwise inhibition by all its hydroxylated metabolites, which include 2-hydroxy-FYX-051, dihydroxy-FYX-051, and trihydroxy-FYX-051. However, the detailed mechanism of inhibition of each metabolite remains elusive and can be useful for designing more effective drugs with similar inhibition functions. Hence, herein we present the computational investigation of the structural and dynamical effects of FYX-051 and the calculated reaction mechanism for all of the oxidation steps catalyzed by the molybdopterin center in the active site. Calculated results for the proposed reaction mechanisms for each metabolite's inhibition reaction in the enzyme's active site, binding affinities, and the noncovalent interactions with the surrounding amino acid residues are consistent with previously reported experimental findings. Analysis of the noncovalent interactions via energy decomposition analysis (EDA) and noncovalent interaction (NCI) techniques suggests that residues L648, K771, E802, R839, L873, R880, R912, F914, F1009, L1014, and A1079 can be used as key interacting residues for further hybrid-type inhibitor development.
Collapse
Affiliation(s)
- Yazdan Maghsoud
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Chao Dong
- Department of Chemistry and Physics, The University of Texas Permian Basin, Odessa, Texas 79762, United States
| | - G Andrés Cisneros
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States; Department of Physics, The University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
9
|
Yang KJ, Choi WJ, Chang YK, Park CW, Kim SY, Hong YA. Inhibition of Xanthine Oxidase Protects against Diabetic Kidney Disease through the Amelioration of Oxidative Stress via VEGF/VEGFR Axis and NOX-FoxO3a-eNOS Signaling Pathway. Int J Mol Sci 2023; 24:ijms24043807. [PMID: 36835220 PMCID: PMC9961241 DOI: 10.3390/ijms24043807] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/05/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Xanthine oxidase (XO) is an important source of reactive oxygen species. This study investigated whether XO inhibition exerts renoprotective effects by inhibiting vascular endothelial growth factor (VEGF) and NADPH oxidase (NOX) in diabetic kidney disease (DKD). Febuxostat (5 mg/kg) was administered to streptozotocin (STZ)-treated 8-week-old male C57BL/6 mice via intraperitoneal injection for 8 weeks. The cytoprotective effects, its mechanism of XO inhibition, and usage of high-glucose (HG)-treated cultured human glomerular endothelial cells (GECs) were also investigated. Serum cystatin C, urine albumin/creatinine ratio, and mesangial area expansion were significantly improved in febuxostat-treated DKD mice. Febuxostat reduced serum uric acid, kidney XO levels, and xanthine dehydrogenase levels. Febuxostat suppressed the expression of VEGF mRNA, VEGF receptor (VEGFR)1 and VEGFR3, NOX1, NOX2, and NOX4, and mRNA levels of their catalytic subunits. Febuxostat caused downregulation of Akt phosphorylation, followed by the enhancement of dephosphorylation of transcription factor forkhead box O3a (FoxO3a) and the activation of endothelial nitric oxide synthase (eNOS). In an in vitro study, the antioxidant effects of febuxostat were abolished by a blockade of VEGFR1 or VEGFR3 via NOX-FoxO3a-eNOS signaling in HG-treated cultured human GECs. XO inhibition attenuated DKD by ameliorating oxidative stress through the inhibition of the VEGF/VEGFR axis. This was associated with NOX-FoxO3a-eNOS signaling.
Collapse
Affiliation(s)
- Keum-Jin Yang
- Clinical Research Institute, Daejeon St. Mary’s Hospital, 64, Daeheung-ro, Jung-gu, Daejeon 34943, Republic of Korea
| | - Won Jung Choi
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yoon-Kyung Chang
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Cheol Whee Park
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Suk Young Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yu Ah Hong
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- Correspondence: ; Tel.: +82-42-220-9255
| |
Collapse
|
10
|
Synthesis, characterization and inhibitor properties of benzimidazolium salts bearing 4-(methylsulfonyl)benzyl side arms. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
11
|
Noda M, Kikuchi C, Tarui R, Nakamura T, Murase T, Hori E, Matsunaga T. Effect of Topiroxostat on Reducing Oxidative Stress in the Aorta of Streptozotocin-Induced Diabetic Rats. Biol Pharm Bull 2023; 46:272-278. [PMID: 36529499 DOI: 10.1248/bpb.b22-00694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Xanthine oxidoreductase exists both intracellularly and extracellularly and induces vascular injury by producing reactive oxygen species (ROS). Here, we investigated the effects and mechanism of action of topiroxostat, a xanthine oxidase inhibitor, on ROS using an animal model of type 1 diabetes with persistent hyperglycemia. Six-week-old male Sprague-Dawley rats were administered 50 mg/kg streptozotocin to induce diabetes; at 8 weeks of age, animals were administered topiroxostat (0.3, 1, or 3 mg/kg) for 2 weeks through mixed feeding after which the aorta was sampled. The production of superoxide, a type of ROS, was measured by chemiluminescence and dihydroethidium staining. Cytotoxicity was evaluated by nitrotyrosine staining. Topiroxostat at 3 mg/kg significantly decreased blood urea nitrogen, e-selectin, urinary malondialdehyde, and the urinary albumin/creatinine ratio compared with the streptozotocin group. Superoxide production by xanthine oxidase anchored to the cell membrane was significantly decreased by topiroxostat at both 1 mg/kg and 3 mg/kg compared with the streptozotocin group. Dihydroethidium staining revealed no significant effect of topiroxostat administration on superoxide production. The fluorescence intensity of nitrotyrosine staining was significantly suppressed by 3 mg/kg topiroxostat. Topiroxostat was found to inhibit the production of ROS in the thoracic aorta and suppress vascular endothelial damage. The antioxidant effect of topiroxostat appears to be exerted via the inhibition of anchored xanthine oxidase.
Collapse
Affiliation(s)
- Masato Noda
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Chigusa Kikuchi
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University.,Laboratory of Community Medicine, Showa Pharmaceutical University.,Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University
| | - Ryota Tarui
- Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University
| | - Takashi Nakamura
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho
| | - Takayo Murase
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho
| | - Eisei Hori
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University.,Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University.,Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
12
|
Kaya G, Noma SAA, Barut Celepci D, Bayıl İ, Taskin-Tok T, Gök Y, Ateş B, Aktaş A, Aygün M, Tezcan B. Design, synthesis, spectroscopic characterizations, single crystal X-ray analysis, in vitro xanthine oxidase and acetylcholinesterase inhibitory evaluation as well as in silico evaluation of selenium-based N-heterocyclic carbene compounds. J Biomol Struct Dyn 2023; 41:11728-11747. [PMID: 36622368 DOI: 10.1080/07391102.2022.2163696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/22/2022] [Indexed: 01/10/2023]
Abstract
Herein, eight new NHC-based selenourea derivatives were synthesized and characterized by using spectroscopic method (1H, 19F, and 13C NMR, FT-IR), and elemental analysis techniques. These compounds were synthesized by mixing benzimidazolium salts, potassium carbonate, and selenium powder in ethyl alcohol. Additionally, the molecular and crystal structures of the three compounds (1c, 2b, and 2c) were determined using the single-crystal x-ray diffraction (XRD) method. Diffraction analysis demonstrated the partial carbon-selenium double-bond character of these compounds. All compounds were determined to be highly potent inhibitors for AChE and XO enzymes. The IC50 values for the compounds were found in the range of 0.361-0.754 μM for XO and from 0.995 to 1.746 μM for AChE. The DNA binding properties of the compounds were investigated. These compounds did not have a remarkable DNA binding property. Also, DPPH radical scavenging activities of the compounds were also investigated. Compounds (1c), (2a), (3a), and (3b) exhibited more pronounced DPPH radical scavenging activity when compared to other compounds. Docking studies were applied by using AutoDock 4 to determine interaction mechanism of the selected compounds (1a), (1b), and (3b). The compound (1b) has good binding affinity (-9.78 kcal/mol) against AChE, and (-6.86 kcal/mol) for XO target. Drug similarity properties of these compounds compared to positive controls were estimated and evaluated by ADMET analysis. Furthermore, molecular dynamics simulations have been applied to understand the accuracy of docking studies. These findings and the defined compounds could be potential candidates for the discovery and progress of effective medicine(s) for AChE and XO in the future.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Gülşen Kaya
- Department of Chemistry, Faculty of Arts and Science, Inonu University, Malatya, Türkiye
| | - Samir Abbas Ali Noma
- Department of Chemistry, Faculty of Arts and Science, Inonu University, Malatya, Türkiye
- Department of Chemistry, Faculty of Arts and Science, Bursa Uludag University, Bursa, Türkiye
| | - Duygu Barut Celepci
- Faculty of Science, Department of Physics, Dokuz Eylül University, Buca, İzmir, Türkiye
| | - İmren Bayıl
- Department of Bioinformatics and Computational Biology, Institute of Health Sciences, Gaziantep University, Gaziantep, Türkiye
| | - Tugba Taskin-Tok
- Department of Chemistry, Faculty of Arts and Sciences, Gaziantep University, Gaziantep, Türkiye
- Department of Bioinformatics and Computational Biology, Institute of Health Sciences, Gaziantep University, Gaziantep, Türkiye
| | - Yetkin Gök
- Department of Chemistry, Faculty of Arts and Science, Inonu University, Malatya, Türkiye
| | - Burhan Ateş
- Department of Chemistry, Faculty of Arts and Science, Inonu University, Malatya, Türkiye
| | - Aydın Aktaş
- Vocational School of Health Service, Inonu University, Malatya, Türkiye
| | - Muhittin Aygün
- Faculty of Science, Department of Physics, Dokuz Eylül University, Buca, İzmir, Türkiye
| | - Burcu Tezcan
- Department of Chemistry, Faculty of Arts and Science, Cukurova University, Adana, Türkiye
| |
Collapse
|
13
|
Nishino M, Egami Y, Kawanami S, Sugae H, Ukita K, Kawamura A, Nakamura H, Matsuhiro Y, Yasumoto K, Tsuda M, Okamoto N, Matsunaga-Lee Y, Yano M, Tanouchi J, Yamada T, Yasumura Y, Tamaki S, Hayashi T, Nakagawa A, Nakagawa Y, Sotomi Y, Nakatani D, Hikoso S, Sakata Y. Lowering Uric Acid May Improve Prognosis in Patients With Hyperuricemia and Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2022; 11:e026301. [PMID: 36129035 PMCID: PMC9673694 DOI: 10.1161/jaha.122.026301] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background An association between uric acid (UA) and cardiovascular diseases, including heart failure (HF), has been reported. However, whether UA is a causal risk factor for HF is controversial. In particular, the prognostic value of lowering UA in patients with HF with preserved ejection fraction (HFpEF) is unclear. Methods and Results We enrolled patients with HFpEF from the PURSUIT‐HFpEF (Prospective Multicenter Observational Study of Patients With Heart Failure With Preserved Ejection Fraction) registry. We investigated whether UA was correlated with the composite events, including all‐cause mortality and HF rehospitalization, in patients with hyperuricemia and HFpEF (UA >7.0 mg/dL). Additionally, we evaluated whether lowering UA for 1 year (≥1.0 mg/dL) in them reduced mortality or HF rehospitalization. We finally analyzed 464 patients with hyperuricemia. In multivariable Cox regression analysis, UA was an independent determinant of composite death and rehospitalization (hazard ratio [HR], 1.15 [95% CI, 1.03–1.27], P=0.015). We divided them into groups with severe and mild hyperuricemia according to median estimated value of serum UA (8.3 mg/dL). Cox proportional hazards models revealed the incidence of all‐cause mortality was significantly higher in the group with severe hyperuricemia than in the group with mild hyperuricemia (HR, 1.73 [95% CI, 1.19–2.25], P=0.004). The incidence of all‐cause mortality was significantly decreased in the group with lowering UA compared with the group with nonlowering UA (HR, 1.71 [95% CI, 1.02–2.86], P=0.041). The incidence of urate‐lowering therapy tended to be higher in the group with lowering UA than in the group with nonlowering UA (34.9% versus 24.6%, P=0.06). Conclusions UA is a predictor for the composite of all‐cause death and HF rehospitalization in patients with hyperuricemia and HFpEF. In these patients, lowering UA, including the use of urate‐lowering therapy, may improve prognosis.
Collapse
Affiliation(s)
| | | | | | - Hiroki Sugae
- Division of Cardiology Osaka Rosai Hospital Osaka Japan
| | - Kohei Ukita
- Division of Cardiology Osaka Rosai Hospital Osaka Japan
| | | | | | | | - Koji Yasumoto
- Division of Cardiology Osaka Rosai Hospital Osaka Japan
| | - Masaki Tsuda
- Division of Cardiology Osaka Rosai Hospital Osaka Japan
| | | | | | | | - Jun Tanouchi
- Division of Cardiology Osaka Rosai Hospital Osaka Japan
| | - Takahisa Yamada
- Division of Cardiology Osaka General Medical Center Osaka Japan
| | | | - Shunsuke Tamaki
- Division of Cardiology Rinku General Medical Center Osaka Japan
| | | | - Akito Nakagawa
- Division of Cardiology Amagasaki Chuo Hospital Hyogo Japan.,Department of Medical Informatics Osaka University Graduate School of Medicine Suita Japan
| | - Yusuke Nakagawa
- Division of Cardiology Kawanishi City Hospital Kawanishi Japan
| | - Yohei Sotomi
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Daisaku Nakatani
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Shungo Hikoso
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine Osaka University Graduate School of Medicine Suita Japan
| | | |
Collapse
|
14
|
Novel Antibacterial Activity of Febuxostat, an FDA-Approved Antigout Drug against Mycobacterium tuberculosis Infection. Antimicrob Agents Chemother 2022; 66:e0076222. [PMID: 36040172 PMCID: PMC9487535 DOI: 10.1128/aac.00762-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Accumulating evidence suggests that drug repurposing has drawn attention as an anticipative strategy for controlling tuberculosis (TB), considering the dwindling drug discovery and development pipeline. In this study, we explored the antigout drug febuxostat and evaluated its antibacterial activity against Mycobacterium species. Based on MIC evaluation, we found that febuxostat treatment significantly inhibited mycobacterial growth, especially that of Mycobacterium tuberculosis (Mtb) and its phylogenetically close neighbors, M. bovis, M. kansasii, and M. shinjukuense, but these microorganisms were not affected by allopurinol and topiroxostat, which belong to a similar category of antigout drugs. Febuxostat concentration-dependently affected Mtb and durably mediated inhibitory functions (duration, 10 weeks maximum), as evidenced by resazurin microtiter assay, time-kill curve analysis, phenotypic susceptibility test, and the Bactec MGIT 960 system. Based on these results, we determined whether the drug shows antimycobacterial activity against Mtb inside murine bone marrow-derived macrophages (BMDMs). Notably, febuxostat markedly suppressed the intracellular growth of Mtb in a dose-dependent manner without affecting the viability of BMDMs. Moreover, orally administered febuxostat was efficacious in a murine model of TB with reduced bacterial loads in both the lung and spleen without the exacerbation of lung inflammation, which highlights the drug potency. Taken together, unexpectedly, our data demonstrated that febuxostat has the potential for treating TB.
Collapse
|
15
|
Yang YS, Wang B, Zhou KM, Liu J, Jiao QC, Qin P. Discovery of derivatives from Spartina alterniflora-sourced moiety as xanthine oxidase inhibitors to lower uric acid. Bioorg Med Chem Lett 2022; 73:128907. [PMID: 35902063 DOI: 10.1016/j.bmcl.2022.128907] [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: 06/04/2022] [Revised: 07/16/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
In this work, hit compounds Spartinin F1-F20 sharing the Spartina alterniflora-sourced ferulic acid backbone were synthesized and evaluated on inhibiting xanthine oxidase and lowering uric acid level. The top hit Spartinin F2 exhibited inhibition percentages at 10 μM dosage as high as 84.48 (higher than that of the positive control allopurinol) and low cyto-toxicity. Spartinin F2 inferred potential efficiency in lowering the serum UA level (from 631.6 μM to 295.0 μM), which was comparable with allopurinol (to 309.2 μM). Spartinin F2 was also beneficial for other serum indexes. The bioavailability of Spartinin F2 was 63.71% from the preliminary pharmacokinetics test and the molecular docking simulation indicated that except for retaining the hydrogen bonds with the key residues such as THR 1010 and LYS 771, the introduction of the π-sulfur interactions via the sulfonate might also be beneficial for developing more potent XO inhibitors.
Collapse
Affiliation(s)
- Yu-Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Research and Development Center, Nanjing Shibeitai Biotechnology Co., Ltd., Nanjing 210003, China.
| | - Bin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Kang-Min Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Junzhong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Qin-Cai Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Pei Qin
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China; Research and Development Center, Nanjing Shibeitai Biotechnology Co., Ltd., Nanjing 210003, China.
| |
Collapse
|
16
|
Nishino M, Egami Y, Nakamura H, Ukita K, Kawamura A, Matsuhiro Y, Yasumoto K, Tsuda M, Tanaka A, Okamoto N, Matsunaga‐Lee Y, Yano M, Shutta R, Tanouchi J. Prospective randomized comparison of effect on coronary endothelial and renal function between febuxostat and benzbromarone in hyperuricemic patients with coronary artery disease: EFEF study. Health Sci Rep 2022; 5:e563. [PMID: 35356803 PMCID: PMC8939499 DOI: 10.1002/hsr2.563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/01/2022] [Accepted: 02/06/2022] [Indexed: 11/08/2022] Open
Abstract
Background and Aims There are two types of serum uric acid-lowering agents, the xanthine oxidoreductase (XO) inhibitor and non-XO inhibitor. We investigated whether febuxostat, XO inhibitor, could produce more favorable effects on coronary endothelial function (CEF) and renal function than benzbromarone, non-XO inhibitor, in hyperuricemic coronary artery disease (CAD) patients. Methods We divided 21 hyperuricemic patients with stenting for left anterior descending (LAD) or left circumflex (LCX) artery into patients started on febuxostat (F group) and those on benzbromarone (B group). After 8 months, all patients underwent CEF evaluations (acetylcholine provocation test) and optical coherence tomography (OCT) for non-culprit vessels (e.g. if patients received LAD stenting, we evaluated LCX). We compared the diameter ratio induced by acetylcholine and baseline (CEF ratio), thin-cap fibroatheroma and calcified plaque by OCT, uric acid, oxidative stress biomarkers, and renal function including estimated glomerular filtration rate (eGFR) between F and B groups. Creatinine 2 days after stenting was measured to evaluate contrast-induced nephropathy (CIN). Results Change of eGFR was significantly lower in F group (n= 11) than B group over 8 months while the other parameters including CEF ratio were similar. F group showed favorable effects for CIN. Conclusion In conclusion, 8-months of febuxostat, XO inhibitor, does not significantly protect CEF but can protect the renal function including CIN in hyperuricemic patients with CAD compared to benzbromarone, non-XO inhibitor.
Collapse
Affiliation(s)
| | | | | | - Kohei Ukita
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| | | | | | - Koji Yasumoto
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| | - Masaki Tsuda
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| | | | | | | | | | - Ryu Shutta
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| | - Jun Tanouchi
- Division of CardiologyOsaka Rosai HospitalOsakaJapan
| |
Collapse
|
17
|
Chandrashekara S, Paul B. A narrative review of clinical evidence validating the efficacy of topiroxostat in managing hyperuricemia. INDIAN JOURNAL OF RHEUMATOLOGY 2022. [DOI: 10.4103/injr.injr_59_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
18
|
Wakita M, Asai K, Kubota Y, Koen M, Shimizu W. Effect of Topiroxostat on Brain Natriuretic Peptide Level in Patients with Heart Failure with Preserved Ejection Fraction: A Pilot Study. J NIPPON MED SCH 2021; 88:423-431. [PMID: 33455978 DOI: 10.1272/jnms.jnms.2021_88-518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Various optimal medical therapies have been established to treat heart failure (HF) with reduced ejection fraction (HFrEF). Both HFrEF and HF with preserved ejection fraction (HFpEF) are associated with poor outcomes. We investigated the effect of topiroxostat, an oral xanthine oxidoreductase inhibitor, for HFpEF patients with hyperuricemia or gout. METHODS In this nonrandomized, open-label, single-arm trial, we administered topiroxostat 40-160 mg/day to HFpEF patients with hyperuricemia or gout to achieve a target uric acid level of 6.0 mg/dL. The primary outcome was rate of change in log-transformed brain natriuretic peptide (BNP) level from baseline to 24 weeks after topiroxostat treatment. The secondary outcomes included amount of change in BNP level, uric acid evaluation values, and oxidative stress marker levels after 24 weeks of topiroxostat treatment. Thirty-six patients were enrolled; three were excluded before study initiation. RESULTS Change in log-transformed BNP level was -3.4 ± 8.9% (p = 0.043) after 24 weeks of topiroxostat treatment. The rate of change for the decrease in BNP level was -18.0 (-57.7, 4.0 pg/mL; p = 0.041). Levels of uric acid and 8-hydroxy-2'-deoxyguanosine/creatinine, an oxidative stress marker, also significantly decreased (-2.8 ± 1.6 mg/dL, p < 0.001, and -2.3 ± 3.7 ng/mgCr, p = 0.009, respectively). CONCLUSIONS BNP level was significantly lower in HFpEF patients with hyperuricemia or gout after topiroxostat administration; however, the rate of decrease was low. Further trials are needed to confirm our findings.
Collapse
Affiliation(s)
- Masaki Wakita
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Kuniya Asai
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Yoshiaki Kubota
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Masahiro Koen
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School
| |
Collapse
|
19
|
Tsukamoto S, Okami N, Yamada T, Azushima K, Yamaji T, Kinguchi S, Uneda K, Kanaoka T, Wakui H, Tamura K. Prevention of kidney function decline using uric acid-lowering therapy in chronic kidney disease patients: a systematic review and network meta-analysis. Clin Rheumatol 2021; 41:911-919. [PMID: 34642880 DOI: 10.1007/s10067-021-05956-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Several previous studies have suggested that uric acid-lowering therapy (ULT) can slow the progression of chronic kidney disease (CKD). Although crucial for CKD patients, few studies have evaluated the effects of different ULT medications on kidney function. This systematic review summarizes evidence from randomized controlled trials (RCTs) regarding the effects of ULT on kidney function. METHOD We performed a systematic search of PubMed, MEDLINE, Embase, Scopus, and the Cochrane Library up to September 2021 to identify RCTs in CKD patients comparing the effects of ULT on kidney function with other ULT medications or placebo. A network meta-analysis was performed to compare each ULT indirectly. The primary outcome was a change in estimated glomerular filtration rate (eGFR) from baseline. RESULTS Ten studies were selected with a total of 1480 patients. Topiroxostat significantly improved eGFR and reduced the urinary albumin/creatinine ratio compared to placebo (mean difference (MD) and 95% confidence interval [95% CI]: 1.49 [0.08; 2.90], P = 0.038 and 25.65% [13.25; 38.04], P < 0.001, respectively). Although febuxostat did not show a positive effect overall, it significantly improved renal function (i.e., eGFR) in a subgroup of CKD patients with hyperuricemia (MD [95% CI]: 0.85 [0.02; 1.67], P = 0.045). Allopurinol and pegloticase did not show beneficial effects. CONCLUSIONS Topiroxostat and febuxostat may have better renoprotective effects in CKD patients than other ULT medications. Further large-scale, long-term studies are required to determine whether these effects will lead, ultimately, to reductions in dialysis induction and major adverse cardiovascular events. Key Points • This study is the first network meta-analysis comparing the nephroprotective effects of ULT in CKD patients. • Topiroxostat and febuxostat showed better renoprotective effects in CKD patients than other ULT medications. • Heterogeneity was low in this study, suggesting consistency of results.
Collapse
Affiliation(s)
- Shunichiro Tsukamoto
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Naohito Okami
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takayuki Yamada
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takahiro Yamaji
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kazushi Uneda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Department of Kampo Medicine, Aizu Medical Center, Fukushima Medical University School of Medicine, Aizuwakamatsu, Japan
| | - Tomohiko Kanaoka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
20
|
Yılmaz Ü, Noma SAA, Taşkın Tok T, Şen B, Gök Y, Aktaş A, Ateş B, Aygün M. A study about excellent xanthine oxidase inhibitory effects of new pyridine salts. MONATSHEFTE FUR CHEMIE 2021. [DOI: 10.1007/s00706-021-02831-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
21
|
Kawachi Y, Fujishima Y, Nishizawa H, Nakamura T, Akari S, Murase T, Saito T, Miyazaki Y, Nagao H, Fukuda S, Kita S, Katakami N, Doki Y, Maeda N, Shimomura I. Increased plasma XOR activity induced by NAFLD/NASH and its possible involvement in vascular neointimal proliferation. JCI Insight 2021; 6:e144762. [PMID: 34494551 PMCID: PMC8492303 DOI: 10.1172/jci.insight.144762] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 07/21/2021] [Indexed: 12/20/2022] Open
Abstract
Xanthine oxidoreductase (XOR) is an enzyme that catalyzes hypoxanthine to xanthine and xanthine to uric acid, respectively. However, the underlying mechanisms of increased plasma XOR and its pathological roles in systemic diseases, such as atherosclerosis, are not fully understood. In this study, we found that changes in plasma XOR activity after bariatric surgery closely associated with those in liver enzymes, but not with those in BMI. In a mouse model of nonalcoholic fatty liver disease/steatohepatitis (NAFLD/NASH), plasma XOR activity markedly increased. Besides, purine catabolism was accelerated in the plasma per se of NASH mice and human patients with high XOR activity. In our NASH mice, we observed an increased vascular neointima formation consisting of dedifferentiated vascular smooth muscle cells (SMCs), which was significantly attenuated by topiroxostat, a selective XOR inhibitor. In vitro, human liver S9–derived XOR promoted proliferation of SMCs with phenotypic modulation and induced ROS production by catabolizing hypoxanthine released from human endothelial cells. Collectively, the results from human and mouse models suggest that increased plasma XOR activity, mainly explained by excess hepatic leakage, was involved in the pathogenesis of vascular injury, especially in NAFLD/NASH conditions.
Collapse
Affiliation(s)
- Yusuke Kawachi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuya Fujishima
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | - Seigo Akari
- Sanwa Kagaku Kenkyusho Co., Ltd., Inabe, Mie, Japan
| | | | | | | | - Hirofumi Nagao
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shiro Fukuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Adipose Management, and
| | - Naoto Katakami
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
22
|
Zhang Y, Fukusumi Y, Kayaba M, Nakamura T, Sakamoto R, Ashizawa N, Kawachi H. Xanthine oxidoreductase inhibitor topiroxostat ameliorates podocyte injury by inhibiting the reduction of nephrin and podoplanin. Nefrologia 2021; 41:539-547. [PMID: 36165136 DOI: 10.1016/j.nefroe.2021.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/17/2020] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Topiroxostat, an inhibitor of xanthine oxidoreductase (XOR) was shown to reduce urinary albumin excretion of hyperuricemic patients with chronic kidney disease. However, its pharmacological mechanism is not well understood. In this study, we examined the effects of topiroxostat on glomerular podocytes. Podocyte is characterized by foot process and a unique cell-cell junction slit diaphragm functioning as a final barrier to prevent proteinuria. METHODS The effects of topiroxostat on the expressions of podocyte functional molecules were analysed in db/db mice, a diabetic nephropathy model, anti-nephrin antibody-induced rat podocyte injury model and cultured podocytes treated with adriamycin. RESULTS Topiroxostat treatment ameliorated albuminuria in db/db mice. The expression of desmin, a podocyte injury marker was increased, and nephrin and podocin, key molecules of slit diaphragm, and podoplanin, an essential molecule in maintaining foot process were downregulated in db/db mice. Topiroxostat treatment prevented the alterations in the expressions of these molecules in db/db mice. XOR activity in kidney was increased in rats with anti-nephrin antibody-induced podocyte injury. Topiroxostat treatment reduced XOR activity and restored the decreased expression of nephrin, podocin and podoplanin in the podocyte injury. Furthermore, topiroxostat enhanced the expression of podoplanin in injured human cultured podocytes. CONCLUSIONS Podocyte injury was evident in db/db mice. Topiroxostat ameliorated albuminuria in diabetic nephropathy model by preventing podocyte injury. Increase of XOR activity in kidney contributes to development of podocyte injury caused by stimulation to slit diaphragm. Topiroxostat has an effect to stabilize slit diaphragm and foot processes by inhibiting the reduction of nephrin, podocin and podoplanin.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshiyasu Fukusumi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Mutsumi Kayaba
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takashi Nakamura
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho, Mie, Japan
| | - Ryusuke Sakamoto
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho, Mie, Japan
| | - Naoki Ashizawa
- Biological Research Group, Research Department, Medical R&D Division, Fuji Yakuhin, Saitama, Japan
| | - Hiroshi Kawachi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
23
|
Independent association of plasma xanthine oxidoreductase activity with hypertension in nondiabetic subjects not using medication. Hypertens Res 2021; 44:1213-1220. [PMID: 34117403 DOI: 10.1038/s41440-021-00679-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 02/05/2023]
Abstract
Xanthine oxidoreductase (XOR), a rate-limiting and catalyzing enzyme of uric acid formation in purine metabolism, is involved in reactive oxygen species generation. Plasma XOR activity has been shown to be a novel metabolic biomarker related to obesity, liver dysfunction, hyperuricemia, dyslipidemia, and insulin resistance. However, the association between plasma XOR activity and hypertension has not been fully elucidated. We investigated the association of hypertension with plasma XOR activity in 271 nondiabetic subjects (male/female: 119/152) who had not taken any medications in the Tanno-Sobetsu Study, a population-based cohort. Males had higher plasma XOR activity than females. Plasma XOR activity was positively correlated with mean arterial pressure (r = 0.128, P = 0.036). When the subjects were divided by the presence and absence of hypertension into an HT group (male/female: 34/40) and a non-HT group (male/female: 85/112), plasma XOR activity in the HT group was significantly higher than that in the non-HT group (median: 39 vs. 28 pmol/h/mL, P = 0.028). There was no significant difference in uric acid levels between the two groups. Multivariable logistic regression analysis showed that plasma XOR activity (odds ratio: 1.091 [95% confidence interval: 1.023-1.177] per 10 pmol/h/mL, P = 0.007) was an independent determinant of the risk for hypertension after adjustment for age, sex, current smoking and alcohol consumption, estimated glomerular filtration rate, brain natriuretic peptide, and insulin resistance index. The interaction of sex with plasma XOR activity was not significant for the risk of hypertension. In conclusion, plasma XOR activity is independently associated with hypertension in nondiabetic individuals who are not taking any medications.
Collapse
|
24
|
Fu H, Zhang J, Huang M. Topiroxostat ameliorates oxidative stress and inflammation in sepsis-induced lung injury. ACTA ACUST UNITED AC 2021; 75:425-431. [PMID: 32589612 DOI: 10.1515/znc-2020-0074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/01/2020] [Indexed: 11/15/2022]
Abstract
Sepsis-induced lung injury was the most common cause of death in patients. Topiroxostat, a novel xanthine oxidoreductase inhibitors, possessed obvious organ protectives effects. Xanthine oxidase played a vital role in acute lung injury. The study aimed to investigate the roles of Topiroxostat in sepsis-induced lung injury. The sepsis rats were established using cecum ligation and perforation. The lung damage induced by sepsis was evaluated by Hematoxylin and Eosin staining and lung tissue wet to dry ratio. The oxidative stress was detected by measurement of reactive oxygen species, malondialdehyde, myeloperoxidase and superoxide dismutase (SOD). The pro-inflammatory mediators, tumor necrosis factor-α, interleukin (IL)-1β, IL-6 and monocyte chemotactic protein 1, were measured by Enzyme-Linked Immunosorbent Assay. The cell apoptosis in lung was detected by TUNNEL staining and western blot analysis of apoptosis-related proteins including pro-apoptosis proteins, Bax, cleaved caspase9, cleaved caspase3 and anti-apoptosis protein Bcl2. The results showed that Topiroxostat significantly reduced lung damage, along with decreased oxidative stress, inflammation response and apoptosis in sepsis rats. Topiroxostat exerted markedly protective effects in sepsis-induced lung injury and could be an antioxidant in treating sepsis-induced lung injury.
Collapse
Affiliation(s)
- Haiying Fu
- Emergency Department, Tong Ren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjie Zhang
- Emergency Department, Tong Ren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mayu Huang
- Emergency Department, Tong Ren Hospital Shanghai Jiao Tong University School of Medicine, 1111 Xian xia Road Changning District, Shanghai, China
| |
Collapse
|
25
|
Hydrogen Gas Inhalation Attenuates Endothelial Glycocalyx Damage and Stabilizes Hemodynamics in a Rat Hemorrhagic Shock Model. Shock 2021; 54:377-385. [PMID: 32804466 PMCID: PMC7458091 DOI: 10.1097/shk.0000000000001459] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Supplemental Digital Content is available in the text Background: Hydrogen gas (H2) inhalation during hemorrhage stabilizes post-resuscitation hemodynamics, improving short-term survival in a rat hemorrhagic shock and resuscitation (HS/R) model. However, the underlying molecular mechanism of H2 in HS/R is unclear. Endothelial glycocalyx (EG) damage causes hemodynamic failure associated with HS/R. In this study, we tested the hypothesis that H2 alleviates oxidative stress by suppressing xanthine oxidoreductase (XOR) and/or preventing tumor necrosis factor-alfa (TNF-α)-mediated syndecan-1 shedding during EG damage. Methods: HS/R was induced in rats by reducing mean arterial pressure (MAP) to 35 mm Hg for 60 min followed by resuscitation. Rats inhaled oxygen or H2 + oxygen after achieving shock either in the presence or absence of an XOR inhibitor (XOR-I) for both the groups. In a second test, rats received oxygen alone or antitumor necrosis factor (TNF)-α monoclonal antibody with oxygen or H2. Two hours after resuscitation, XOR activity, purine metabolites, cytokines, syndecan-1 were measured and survival rates were assessed 6 h after resuscitation. Results: H2 and XOR-I both suppressed MAP reduction and improved survival rates. H2 did not affect XOR activity and the therapeutic effects of XOR-I and H2 were additive. H2 suppressed plasma TNF-α and syndecan-1 expression; however, no additional H2 therapeutic effect was observed in the presence of anti-TNF-α monoclonal antibody. Conclusions: H2 inhalation after shock stabilized hemodynamics and improved survival rates in an HS/R model independent of XOR. The therapeutic action of H2 was partially mediated by inhibition of TNF-α-dependent syndecan-1 shedding.
Collapse
|
26
|
Takahashi K, Mizukami H, Osonoi S, Ogasawara S, Hara Y, Kudoh K, Takeuchi Y, Sasaki T, Daimon M, Yagihashi S. Inhibitory effects of xanthine oxidase inhibitor, topiroxostat, on development of neuropathy in db/db mice. Neurobiol Dis 2021; 155:105392. [PMID: 34000416 DOI: 10.1016/j.nbd.2021.105392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation and oxidative stress contribute to the pathophysiology of diabetic neuropathy. According to recent evidence, the modulation of macrophage polarization in peripheral nerves represents a potential therapeutic target for diabetic neuropathy. Xanthine oxidase, which is a form of xanthin oxidoreductase, is the rate-limiting enzyme that catalyzes the degradation of hypoxanthine and xanthine into uric acid. Activation of xanthine oxidase promotes oxidative stress and macrophage activation. A preclinical study reported the beneficial effects of xanthine oxidase inhibitors on peripheral nerve dysfunction in experimental models of diabetes. However, the detailed mechanisms remain unknown. In this study, we examined the effect of the xanthine oxidase inhibitor topiroxostat on macrophage polarization and peripheral neuropathy in an obese diabetic model, db/db mice. First, the effects of xanthine oxidase inhibitors on cultured macrophages and dorsal root ganglion neurons exposed to xanthine oxidase were assessed. Furthermore, five-week-old db/db mice were administered the xanthine oxidase inhibitors topiroxostat [1 mg/kg/day (dbT1) or 2 mg/kg/day (dbT2)] or febuxostat [1 mg/kg (dbF)]. Glucose metabolism and body weight were evaluated during the experimental period. At 4 and 8 weeks of treatment, peripheral nerve functions such as nerve conduction velocities, thermal thresholds and pathology of skin and sciatic nerves were evaluated. The mRNA expression of molecules related to inflammation and oxidative stress was also measured in sciatic nerves. Untreated db/db mice and the nondiabetic db strain (db/m) were studied for comparison. An in vitro study showed that topiroxostat suppressed macrophage activation and proinflammatory but not anti-inflammatory polarization, and prevented the reduction in neurite outgrowth from neurons exposed to xanthine oxidase. Neuropathic changes exemplified by delayed nerve conduction and reduced intraepidermal nerve fiber density developed in db/db mice. These deficits were significantly prevented in the treated group, most potently in dbT2. Protective effects were associated with the suppression of macrophage infiltration, cytokine expression, and oxidative stress in the sciatic nerve and decreased plasma xanthine oxidoreductase activity. Our results revealed the beneficial effects of the xanthine oxidase inhibitor topiroxostat on neuropathy development in a mouse model of type 2 diabetes. The suppression of proinflammatory macrophage activation and oxidative stress-induced damage were suggested to be involved in this process.
Collapse
Affiliation(s)
- Kazuhisa Takahashi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan.
| | - Sho Osonoi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Saori Ogasawara
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Yutaro Hara
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; Department of Gastroenterological Surgery and Pediatric Surgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Kazuhiro Kudoh
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Yuki Takeuchi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Takanori Sasaki
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Makoto Daimon
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Soroku Yagihashi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| |
Collapse
|
27
|
Influence of xanthine oxidoreductase inhibitor, topiroxostat, on body weight of diabetic obese mice. Nutr Diabetes 2021; 11:12. [PMID: 33850106 PMCID: PMC8044114 DOI: 10.1038/s41387-021-00155-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/17/2021] [Accepted: 03/30/2021] [Indexed: 11/09/2022] Open
Abstract
Plasma xanthine oxidoreductase (XOR) activity is high in metabolic disorders such as diabetic mellitus, obesity, or overweight. Thus, this study investigated whether the XOR inhibitor, topiroxostat, affected body weight. Male db/db mice were fed standard diets with or without topiroxostat for 4 weeks. Body weight and food intake were constantly monitored, along with monitoring plasma biochemical markers, including insulin and XOR activity. Additionally, hepatic hypoxanthine and XOR activity were also documented. Single regression analysis was performed to determine the mechanism. Topiroxostat treatment suppressed weight gain relative to the vehicle without any impact on food intake. However, the weight of fat pads and hepatic and muscle triglyceride content did not change. Topiroxostat decreased the plasma uric acid and increased hepatic hypoxanthine in response to the inhibition of XOR activity. Plasma ketone body and free fatty acid were also increased. Moreover, fat weight was weakly associated with plasma XOR activity in the diabetic state and was negatively associated with ketone body by topiroxostat. These results suggested that topiroxostat amplified the burning of lipids and the salvage pathway, resulting in predisposing the body toward catabolism. The inhibition of plasma XOR activity may contribute to weight loss.
Collapse
|
28
|
Li G, Yang H, Liu W, Shen C, Ji Y, Sun Y, Huo Q, Liu Y, Wang G. Development of an In Vivo Predictive Dissolution Methodology of Topiroxostat Immediate-Release Tablet Using In Silico Simulation. AAPS PharmSciTech 2021; 22:132. [PMID: 33851275 DOI: 10.1208/s12249-021-01992-1] [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: 01/13/2021] [Accepted: 03/15/2021] [Indexed: 11/30/2022] Open
Abstract
The main objective of this study was to develop an in vivo predictive dissolution (IVPD) model for topiroxostat immediate-release (IR) formulation by the combination of mechanistic absorption model (MAM) deconvolution method with time shifting factor (TSF) adjustment. The in vitro dissolution profiles in different biorelevant dissolution media containing different concentrations of sodium lauryl sulfate (SLS) were obtained from dissolution testing with the paddle method of the US Pharmacopeia, while the human pharmacokinetic profile was taken from the published experimental results. The GastroPlus™ software was used to observe the linear relationship between in vitro drug dissolution and in vivo absorption. The pharmacokinetic profile of topiroxostat IR tablet was first deconvoluted through the MAM method to obtain the fraction absorbed in vivo. Next, Levy plot was constructed to estimate the TSF, and the time scale for both processes of dissolution and absorption was then adjusted to be superimposable. The IVPD modelling was subsequently established with data between in vitro dissolution profiles and fraction absorbed in vivo. Finally, the dissolution profiles of topiroxostat IR tablet were translated into a pharmacokinetic curve in terms of convolution method. The comparison between translated and observed pharmacokinetic data will validate the performance of the developed IVPD model. This new linear IVPD model with high predictive power for the tablet can predict the in vivo pharmacokinetic differences through in vitro dissolution data, and it can be utilized as a risk-control tool for the formulation development of the topiroxostat IR tablet and the quality control of product batches.
Collapse
|
29
|
Zhang Y, Fukusumi Y, Kayaba M, Nakamura T, Sakamoto R, Ashizawa N, Kawachi H. Xanthine oxidoreductase inhibitor topiroxostat ameliorates podocyte injury by inhibiting the reduction of nephrin and podoplanin. Nefrologia 2021. [PMID: 33707098 DOI: 10.1016/j.nefro.2020.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Topiroxostat, an inhibitor of xanthine oxidoreductase (XOR) was shown to reduce urinary albumin excretion of hyperuricemic patients with chronic kidney disease. However, its pharmacological mechanism is not well understood. In this study, we examined the effects of topiroxostat on glomerular podocytes. Podocyte is characterized by foot process and a unique cell-cell junction slit diaphragm functioning as a final barrier to prevent proteinuria. METHODS The effects of topiroxostat on the expressions of podocyte functional molecules were analysed in db/db mice, a diabetic nephropathy model, anti-nephrin antibody-induced rat podocyte injury model and cultured podocytes treated with adriamycin. RESULTS Topiroxostat treatment ameliorated albuminuria in db/db mice. The expression of desmin, a podocyte injury marker was increased, and nephrin and podocin, key molecules of slit diaphragm, and podoplanin, an essential molecule in maintaining foot process were downregulated in db/db mice. Topiroxostat treatment prevented the alterations in the expressions of these molecules in db/db mice. XOR activity in kidney was increased in rats with anti-nephrin antibody-induced podocyte injury. Topiroxostat treatment reduced XOR activity and restored the decreased expression of nephrin, podocin and podoplanin in the podocyte injury. Furthermore, topiroxostat enhanced the expression of podoplanin in injured human cultured podocytes. CONCLUSIONS Podocyte injury was evident in db/db mice. Topiroxostat ameliorated albuminuria in diabetic nephropathy model by preventing podocyte injury. Increase of XOR activity in kidney contributes to development of podocyte injury caused by stimulation to slit diaphragm. Topiroxostat has an effect to stabilize slit diaphragm and foot processes by inhibiting the reduction of nephrin, podocin and podoplanin.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshiyasu Fukusumi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Mutsumi Kayaba
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takashi Nakamura
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho, Mie, Japan
| | - Ryusuke Sakamoto
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho, Mie, Japan
| | - Naoki Ashizawa
- Biological Research Group, Research Department, Medical R&D Division, Fuji Yakuhin, Saitama, Japan
| | - Hiroshi Kawachi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
30
|
Cicero AFG, Fogacci F, Kuwabara M, Borghi C. Therapeutic Strategies for the Treatment of Chronic Hyperuricemia: An Evidence-Based Update. ACTA ACUST UNITED AC 2021; 57:medicina57010058. [PMID: 33435164 PMCID: PMC7827966 DOI: 10.3390/medicina57010058] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022]
Abstract
This article aims to critically review the evidence on the available therapeutic strategies for the treatment of hyperuricemia. For this reason, several papers were reviewed. Xanthine oxidase inhibitors are the safest and most effective uric acid lowering drugs for the management of chronic hyperuricemia, while the efficacy of uricosuric agents is strongly modulated by pharmacogenetics. Emergent drugs (lesinurad, peglotidase) were found to be more effective for the acute management of refractory hyperuricemia, but their use is supported by a relatively small number of clinical trials so that further well-designed clinical research is needed to deepen their efficacy and safety profile.
Collapse
Affiliation(s)
- Arrigo F. G. Cicero
- Hypertension Research Unit, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.F.G.C.); (F.F.)
| | - Federica Fogacci
- Hypertension Research Unit, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.F.G.C.); (F.F.)
| | - Masanari Kuwabara
- Cardiology Department and Intensive Care Unit, Toranomon Hospital, Tokyo 40138, Japan;
| | - Claudio Borghi
- Hypertension Research Unit, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.F.G.C.); (F.F.)
- Correspondence: ; Tel.: +39-512142224
| |
Collapse
|
31
|
Kario K, Nishizawa M, Kiuchi M, Kiyosue A, Tomita F, Ohtani H, Abe Y, Kuga H, Miyazaki S, Kasai T, Hongou M, Yasu T, Kuramochi J, Fukumoto Y, Hoshide S, Hisatome I. Comparative effects of topiroxostat and febuxostat on arterial properties in hypertensive patients with hyperuricemia. J Clin Hypertens (Greenwich) 2021; 23:334-344. [PMID: 33400348 PMCID: PMC8029836 DOI: 10.1111/jch.14153] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/30/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022]
Abstract
Elevated serum uric acid is a cardiovascular risk factor in patients with hypertension, even when blood pressure (BP) is well controlled. Xanthine oxidoreductase inhibitors (XORi) reduce serum uric acid levels and have several other potential effects. This multicenter, randomized, open‐label study compared the effects of two XORi, topiroxostat and febuxostat, on arterial stiffness, uric acid levels, and BP in hypertensive patients with hyperuricemia. Patients received topiroxostat 40–160 mg/day or febuxostat 10–60 mg/day, titrated to maintain serum uric acid <6 mg/dl, for 24 weeks. The primary endpoint was change in the cardio‐ankle vascular index (CAVI) from baseline to 24 weeks. There were no significant changes in CAVI from baseline to 24 weeks (from 9.13 to 9.16 [feboxustat] and 8.98 to 9.01 [topiroxostat]). Compared with baseline, there were significant reductions in serum uric acid (–2.9 and –2.5 mg/dl; both p < 0.001) and morning home systolic BP (–3.6 and –5.1 mm Hg; both p < 0.01) after 24 weeks' treatment with febuxostat and topiroxostat. BP decreased to the greatest extent in the subgroup of patients with uncontrolled blood pressure at baseline. Topiroxostat, but not febuxostat, significantly decreased plasma xanthine oxidoreductase activity versus baseline. The urinary albumin‐creatinine ratio (UACR) decreased significantly from baseline to 24 weeks with topiroxostat (–20.8%; p = 0.021), but not febuxostat (–8.8%; p = 0.362). In conclusion, neither topiroxostat nor febuxostat had any significant effects on arterial stiffness over 24 weeks' treatment.
Collapse
Affiliation(s)
- Kazuomi Kario
- Jichi Medical University School of Medicine, Tochigi, Japan
| | | | | | | | | | | | | | | | | | | | | | - Takanori Yasu
- Dokkyo Medical University Nikko Medical Center, Tochigi, Japan
| | | | | | | | - Ichiro Hisatome
- Tottori University Graduate School of Medical Science, Tottori, Japan
| |
Collapse
|
32
|
Cicero AFG, Fogacci F, Cincione RI, Tocci G, Borghi C. Clinical Effects of Xanthine Oxidase Inhibitors in Hyperuricemic Patients. Med Princ Pract 2021; 30:122-130. [PMID: 33040063 PMCID: PMC8114083 DOI: 10.1159/000512178] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 10/07/2020] [Indexed: 12/13/2022] Open
Abstract
This review aims to critically present the available clinical evidence supporting the treatment of chronic hyperuricemia with xanthine oxidase inhibitors. For this reason, the studies published on uric acid (UA)-lowering drugs in the English language from 2000 to August 2019 have been carefully reviewed. The terms "serum uric acid," "xanthine oxidase," "allopurinol," "febuxostat," and "topiroxostat" were incorporated into an electronic search strategy, alone and in combinations, in both MEDLINE (National Library of Medicine, Bethesda, MD) and the Cochrane Register of Controlled Trials (The Cochrane Collaboration, Oxford, UK). Even if new urate-lowering drugs seem of particular efficacy for acute treatment of refractory hyperuricemia, their use is supported by relatively small clinical evidence. On the contrary, large long-term clinical trials have demonstrated that xanthine oxidase inhibitors (XOIs, namely, allopurinol and febuxostat) are effective, safe, and relatively well-tolerated in most of the patients. They have mainly been tested in the elderly, in patients affected by chronic diseases such as heart failure and cancer, and in patients taking a large number of drugs, confirming their safety profile. Recent data also show that they could exert some positive effects on vascular health, renal function, and glucose metabolism. Their cost is also low. In conclusion, XOIs remain the first choice of UA-lowering drug for chronic treatment.
Collapse
Affiliation(s)
- Arrigo F G Cicero
- Hypertension Research Unit, Medical and Surgical Sciences Department, University of Bologna, Bologna, Italy,
- IRCCS Policlinico di S.Orsola, Bologna, Italy,
| | - Federica Fogacci
- Hypertension Research Unit, Medical and Surgical Sciences Department, University of Bologna, Bologna, Italy
| | | | - Giuliano Tocci
- Hypertension Unit, Division of Cardiology, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, University of Rome Sapienza, Sant'Andrea Hospital, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Claudio Borghi
- Hypertension Research Unit, Medical and Surgical Sciences Department, University of Bologna, Bologna, Italy
- IRCCS Policlinico di S.Orsola, Bologna, Italy
| |
Collapse
|
33
|
Basnet R, Khadka S, Basnet BB, Basnet TB, Chidi BB, Nirala S, Gupta R, Sharma B. Xanthine Oxidase and Transforming Growth Factor Beta-activated Kinase 1: Potential Targets for Gout Intervention. Curr Rheumatol Rev 2020; 17:153-161. [PMID: 33243128 DOI: 10.2174/1573397116666201126162202] [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: 06/23/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Gout, inflammatory arthritis caused by the deposition of monosodium urate crystals into affected joints and other tissues, has become one of the major health problems of today's world. The main risk factor for gout is hyperuricemia, which may be caused by excessive or insufficient excretion of uric acid. The incidence is usually in the age group of 30- 50 years, commonly in males. In developed countries, the incidence of gout ranges from 1 to 4%. Despite effective treatments, there has been an increase in the number of cases over the past few decades. OBJECTIVE In recent years, the development of targeted drugs in gout has made significant achievements. The global impact of gout continues to increase, and as a result, the focus of disease-modifying therapies remains elusive. In addition, the characterization of available instrumental compounds is urgently needed to explore the use of novel selective and key protein-ligand interactions for the effective treatment of gout. Xanthine oxidase (XO) is a key target in gout to consider the use of XO inhibitors in patients with mild to moderate condition, however, the costs are high, and no other direct progress has been made. Despite many XO inhibitors, a selective potent inhibitor for XO is limited. Likewise, in recent years, attention has been focused on different strategies for the discovery and development of new selectivity ligands against transforming growth factor beta- activated kinase 1 (TAK1), a potential therapeutic target for gout. Therefore, the insight on human XO structure and TAK1 provides a clue into protein-ligand interactions and provides the basis for molecular modeling and structure-based drug design. CONCLUSION In this review, we briefly introduce the clinical characteristics, the development of crystal, inhibitors, and crystal structure of XO and TAK1 protein.
Collapse
Affiliation(s)
- Rajesh Basnet
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Sandhya Khadka
- Department of Pharmacy, Hope International College, Purbanchal University, Lalitpur, Nepal
| | | | - Til Bahadur Basnet
- Little Buddha College of Health Sciences, Purbanchal University, Min Bhawan Kathmandu, Nepal
| | - Buddhi Bal Chidi
- Department of Drug Administration, Government of Nepal, Kathmandu, Nepal
| | - Sanjeev Nirala
- Department of Cardiology of the Ruijin Hospital affiliated to the Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Radheshyam Gupta
- Dept. of General Surgery, Bariatric and Metabolic Surgery, Nepal Korea Friendship Municipality Hospital, Madhyapur Thimi, Nepal, Qiqihar Medical University, Heilongjiang, China
| | - Bidur Sharma
- Department of Pharmacy, School of Science, Kathmandu University, Dhulikhel, Nepal
| |
Collapse
|
34
|
Furuhashi M. New insights into purine metabolism in metabolic diseases: role of xanthine oxidoreductase activity. Am J Physiol Endocrinol Metab 2020; 319:E827-E834. [PMID: 32893671 DOI: 10.1152/ajpendo.00378.2020] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Xanthine oxidoreductase (XOR) consists of two different forms, xanthine dehydrogenase and xanthine oxidase (XO), and is a rate-limiting enzyme of uric acid production from hypoxanthine and xanthine. Uric acid is the end product of purine metabolism in humans and has a powerful antioxidant effect. The lack of ascorbic acid, known as vitamin C, in hominoids has been thought to cause a compensatory increase in uric acid as an antioxidant by unfunctional gene mutation of uricase to a pseudogene. Because XO is involved in an increase in reactive oxygen species (ROS) by generating superoxide and hydrogen peroxide, inadequate activation of XOR promotes oxidative stress-related tissue injury. Plasma XOR activity is associated with obesity, smoking, liver dysfunction, hyperuricemia, dyslipidemia, insulin resistance, and adipokines, indicating a novel biomarker of metabolic disorders. However, XOR activity in adipose tissue is low in humans unlike in rodents, and hypoxanthine is secreted from human adipose tissue. The concentration of hypoxanthine, but not xanthine, is independently associated with obesity in a general population, indicating differential regulation of hypoxanthine and xanthine. Treatment with an XOR inhibitor can decrease uric acid for preventing gout, reduce production of XO-related ROS, and promote reutilization of hypoxanthine and ATP production through the salvage pathway. It has recently been suggested that discontinuation of an XOR inhibitor causes adverse cardiovascular outcomes as XOR inhibitor withdrawal syndrome, possibly due to cardiac disturbance of conduction and contraction by reduced ATP production. New insights into purine metabolism, including the role of XOR activity in the past 5 yr, are mainly discussed in this review.
Collapse
Affiliation(s)
- Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
35
|
Itano S, Kadoya H, Satoh M, Nakamura T, Murase T, Sasaki T, Kanwar YS, Kashihara N. Non-purine selective xanthine oxidase inhibitor ameliorates glomerular endothelial injury in Ins Akita diabetic mice. Am J Physiol Renal Physiol 2020; 319:F765-F772. [PMID: 32954851 DOI: 10.1152/ajprenal.00236.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Endothelial dysfunction represents a predominant early feature of diabetes, rendering patients with diabetes prone to renal complications, e.g., proteinuria. Recent studies have indicated a possible role for xanthine oxidase (XO) in the pathogenesis of vascular dysfunctions associated with diabetes. In the present study, we investigated the contribution of XO activation on the progression of diabetic nephropathy in a mouse model using selective XO inhibitors. Male Ins2Akita heterozygous mice were used with wild-type mice as controls. Akita mice were treated with topiroxostat (Topi) or vehicle for 4 wk. Serum uric acid levels were significantly reduced in Akita + Topi mice compared with Akita + vehicle mice. The Akita + Topi group had a significant reduction in urinary albumin excretion compared with the Akita + vehicle group. Mesangial expansion, glomerular collagen type IV deposition, and glomerular endothelial injury (assessed by lectin staining and transmission electron microscopy) were considerably reduced in the Akita + topi group compared with the Akita + vehicle group. Furthermore, glomerular permeability was significantly higher in the Akita + vehicle group compared with the wild-type group. These changes were reduced with the administration of Topi. We conclude that XO inhibitors preserve glomerular endothelial functions and rescue compromised glomerular permeability, suggesting that XO activation plays a vital role in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Seiji Itano
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Hiroyuki Kadoya
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Minoru Satoh
- Department of General Medicine/Nephrology, Kobe Rosai Hospital, Kobe, Hyogo, Japan
| | - Takashi Nakamura
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho, Mie, Japan
| | - Takayo Murase
- Radioisotope and Chemical Analysis Center, Laboratory Management Department, Sanwa Kagaku Kenkyusho, Mie, Japan
| | - Tamaki Sasaki
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Yashpal S Kanwar
- Department of Pathology and Medicine, Northwestern University, Chicago, Illinois, USA
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| |
Collapse
|
36
|
Feng X, Cao Y, Ding Y, Zheng H. Development and validation for the quantitative determination of xanthine oxidoreductase inhibitor topiroxostat by LC-MS/MS and its clinico-pharmacokinetic study. J Pharm Biomed Anal 2020; 189:113470. [DOI: 10.1016/j.jpba.2020.113470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/22/2020] [Accepted: 07/07/2020] [Indexed: 11/29/2022]
|
37
|
Abstract
Xanthine oxidase inhibitors are primarily used in the clinical prevention and treatment of gout associated with hyperuricemia. The archetypal xanthine oxidase inhibitor, Allopurinol has been shown to have other beneficial effects such as a reduction in vascular reactive oxygen species and mechano-energetic uncoupling. This chapter discusses these properties and their relevance to human pathophysiology with a focus on Allopurinol as well as newer xanthine oxidase inhibitors such as Febuxostat and Topiroxostat. Xanthine oxidase (XO) and xanthine dehydrogenase (XDH) are collectively referred to as xanthine oxidoreductase (XOR). XDH is initially synthesised as a 150-kDa protein from which XO is derived, e.g. under conditions of ischemia/hypoxia either reversibly by conformational changes (calcium or SH oxidation) or irreversibly by proteolysis, the latter leading to formation of a 130-kDa form of XO. Both, XO and XDH, catalyse the conversion of hypoxanthine via xanthine to uric acid, the former by using oxygen forming superoxide and hydrogen peroxide and the latter NAD+. However, XDH is in principle also able to generate ROS.
Collapse
|
38
|
Hosoya T, Uchida S, Shibata S, Tomioka NH, Hosoyamada M. Perfecting a high hypoxanthine phosphoribosyltransferase activity-uricase KO mice to test the effects of purine- and non-purine-type xanthine dehydrogenase (XDH) inhibitors. Br J Pharmacol 2020; 177:2274-2285. [PMID: 31971609 PMCID: PMC7174878 DOI: 10.1111/bph.14978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/25/2019] [Accepted: 12/12/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Purine metabolism in mice and human differ in terms of uricase (Uox) activity as well as hypoxanthine phosphoribosyltransferase (HPRT) activity. The aim of this study was the establishment of high HPRT activity-Uox knockout (KO) mice as a novel hyperuricaemic model. Then to investigate the effects of purine-type xanthine dehydrogenase (XDH) inhibitor, allopurinol, and non-purine-type XDH inhibitor, topiroxostat, on purine metabolism. EXPERIMENTAL APPROACH A novel hyperuricaemic mouse model was established by mating B6-ChrXCMSM mice with uricase KO mice. The pharmacological effects of allopurinol and topiroxostat were explored by evaluating urate, hypoxanthine, xanthine and creatinine in the plasma and urine of these model mice. Furthermore, we analysed the effect of both drugs on erythrocyte hypoxanthine phosphoribosyltransferase activity. KEY RESULTS Plasma urate level and urinary urate/creatinine ratio significantly decreased after administration of allopurinol 30 mg·kg-1 or topiroxostat 1 mg·kg-1 for 7 days. The urate-lowering effect was equivalent for allopurinol and topiroxostat. However, the urinary hypoxanthine/creatinine ratio and xanthine/creatinine ratio after treatment with topiroxostat were significantly lower than for allopurinol. In addition, the urinary oxypurine/creatinine ratio was significantly lowered after treatment with topiroxostat, but allopurinol elicited no such effect. Furthermore, allopurinol inhibited mouse erythrocyte hypoxanthine phosphoribosyltransferase, while topiroxostat did not. CONCLUSIONS AND IMPLICATIONS High hypoxanthine phosphoribosyltransferase activity- uricase KO mice were established as a novel hyperuricaemic animal model. In addition, topiroxostat, a non-purine-type xanthine dehydrogenase inhibitor, elicited a potent plasma urate-lowering effect. However, unlike allopurinol, topiroxostat did not perturb the salvage pathway, resulting in lowered total oxypurine excretion.
Collapse
Affiliation(s)
- Takuji Hosoya
- Department of Human Physiology & Pathology, Faculty of Pharma‐ScienceTeikyo UniversityTokyoJapan
- Biological Research Department, Medical R&D DivisionFuji Yakuhin Co., Ltd.SaitamaJapan
| | - Shunya Uchida
- Division of Nephrology, Department of Internal MedicineTeikyo University School of MedicineTokyoJapan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal MedicineTeikyo University School of MedicineTokyoJapan
| | - Naoko H. Tomioka
- Department of Human Physiology & Pathology, Faculty of Pharma‐ScienceTeikyo UniversityTokyoJapan
| | - Makoto Hosoyamada
- Department of Human Physiology & Pathology, Faculty of Pharma‐ScienceTeikyo UniversityTokyoJapan
| |
Collapse
|
39
|
Morimoto C, Tamura Y, Asakawa S, Kuribayashi-Okuma E, Nemoto Y, Li J, Murase T, Nakamura T, Hosoyamada M, Uchida S, Shibata S. ABCG2 expression and uric acid metabolism of the intestine in hyperuricemia model rat. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2020; 39:744-759. [PMID: 31983315 DOI: 10.1080/15257770.2019.1694684] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To elucidate roles of the intestine in uric acid (UA) metabolism, we examined ABCG2 expression, tissue UA content and xanthine oxidoreductase (XOR) activity in different intestinal segments. Male SD rats were assigned to control group or oxonic acid-induced hyperuricemia (HUA) group. In control rats, ABCG2 was present both in villi and crypts in each segment. Tissue UA content and XOR activity were relatively high in duodenum and jejunum. However, in HUA rats, tissue UA content was significantly elevated in the ileum, whereas it remained unaltered in other segments. Moreover, ABCG2 expression in the HUA group was upregulated both in the villi and crypts of the ileum. These data indicate that the ileum may play an important role in the extra-renal UA excretion.
Collapse
Affiliation(s)
- Chikayuki Morimoto
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshifuru Tamura
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shinichiro Asakawa
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Emiko Kuribayashi-Okuma
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshikazu Nemoto
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Jinping Li
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takayo Murase
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho, Mie, Japan
| | - Takashi Nakamura
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho, Mie, Japan
| | - Makoto Hosoyamada
- Department of Human Physiology and Pathology, Faculty of Pharma-Sciences, Teikyo University, Tokyo, Japan
| | - Shunya Uchida
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan.,Department of Health Care, Teikyo Heisei University, Tokyo, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Matsushita M, Shirakabe A, Okazaki H, Shibata Y, Goda H, Shigihara S, Asano K, Tani K, Kiuchi K, Murase T, Nakamura T, Takayasu T, Asano M, Okajima F, Kobayashi N, Hata N, Asai K, Shimizu W. Plasma Xanthine Oxidoreductase (XOR) Activity in Cardiovascular Disease Outpatients. Circ Rep 2020; 2:104-112. [PMID: 33693215 PMCID: PMC7929762 DOI: 10.1253/circrep.cr-19-0116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background:
The mechanisms of the increased plasma xanthine oxidoreductase (XOR) activity in outpatients with cardiovascular disease were unclear. Methods and Results:
A total of 372 outpatients were screened, and 301 outpatients with cardiovascular disease were prospectively analyzed. Blood samples were collected from patients who visited a daily cardiovascular outpatient clinic. Patients with diabetes mellitus (DM) were significantly more likely to be classified into the high-XOR group (≥100 pg/h/mL; 50%) than the low-XOR group (<100 pmol/h/mL; 28.7%). On multivariate logistic regression analysis, DM (OR, 2.683; 95% CI: 1.441–4.996) was independently associated with high plasma XOR activity in all cohorts. In the diabetic cardiovascular disease patients (n=100), median body mass index (BMI) in the high-XOR group (28.0 kg/m2; IQR, 25.2–29.4 kg/m2, n=32) was significantly higher than in the low-XOR group (23.6 kg/m2; IQR, 21.2–25.7 kg/m2, n=68), and BMI was independently associated with high plasma XOR activity (OR, 1.340; 95% CI: 1.149–1.540). Plasma hydrogen peroxide was significantly higher in DM patients with high plasma XOR activity and obesity (>22 kg/m2) than in other patients. Conclusions:
DM with obesity is one of the mechanisms of XOR enhancement in cardiovascular disease. The increase of XOR is a possible pathway for the production of reactive oxygen species in obese cardiovascular disease patients with DM.
Collapse
Affiliation(s)
- Masato Matsushita
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Akihiro Shirakabe
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Hirotake Okazaki
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Yusaku Shibata
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Hiroki Goda
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Shota Shigihara
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Kazuhiro Asano
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Kenichi Tani
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Kazutaka Kiuchi
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Takayo Murase
- Department of Radioisotope and Chemical Analysis Center, Sanwa Kagaku Kenkyusho Co., Ltd Mie Japan
| | - Takashi Nakamura
- Department Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd Mie Japan
| | - Tsutomu Takayasu
- Department of Internal Medicine, Toho Kamagaya Hospital Chiba Japan
| | - Miwako Asano
- Department of Internal Medicine, Hasegawa Hospital Chiba Japan
| | - Fumitaka Okajima
- Department of Endocrinology, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Nobuaki Kobayashi
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Noritake Hata
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Kuniya Asai
- Division of Intensive Care Unit, Nippon Medical School Chiba Hokusoh Hospital Chiba Japan
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School Tokyo Japan
| |
Collapse
|
41
|
Chen Y, Gao Y, Wu F, Luo X, Ju X, Liu G. Computationally exploring novel xanthine oxidase inhibitors using docking-based 3D-QSAR, molecular dynamics, and virtual screening. NEW J CHEM 2020. [DOI: 10.1039/d0nj03221b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Computationally exploring novel potential xanthine oxidase inhibitors using a systematic modeling study.
Collapse
Affiliation(s)
- Yanming Chen
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Ya Gao
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Fengshou Wu
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Xiaogang Luo
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Xiulian Ju
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Genyan Liu
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| |
Collapse
|
42
|
Prediction of the pharmacokinetics and pharmacodynamics of topiroxostat in humans by integrating the physiologically based pharmacokinetic model with the drug-target residence time model. Biomed Pharmacother 2020; 121:109660. [DOI: 10.1016/j.biopha.2019.109660] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/03/2019] [Accepted: 11/06/2019] [Indexed: 01/12/2023] Open
|
43
|
Matsuo H, Ishikawa E, Machida H, Mizutani Y, Tanoue A, Ohnishi T, Murata T, Okamoto S, Ogura T, Nishimura Y, Ito H, Yasutomi M, Katayama K, Nomura S, Ito M. Efficacy of xanthine oxidase inhibitor for chronic kidney disease patients with hyperuricemia. Clin Exp Nephrol 2019; 24:307-313. [PMID: 31845065 DOI: 10.1007/s10157-019-01829-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/29/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Hyperuricemia is a known risk factor for end-stage renal disease. Although xanthine oxidase (XO) inhibitors are expected to protect the kidney function, evidence to this end is insufficient at present. METHODS This study was a multi-center, open-labeled, randomized study conducted in Mie Prefecture in Japan. Patients were included if they were between 20 and 80 years old and had a serum uric acid (sUA) level ≥ 7.0 mg/dl with or without gout, estimated glomerular filtration rate (eGFR) of 15-60 ml/min/1.73 m2, and urinary protein creatinine ratio (uPCR) of 0.15-3.5 g/gCr. Patients were randomly assigned to a Topiroxostat or Febuxostat group, and the treatment target for the sUA level was < 6.0 mg/dl. The primary outcome was the change in the uPCR after 24 weeks. RESULTS The change in the median uPCR after 24 weeks was not statistically significant after treatment in the Topiroxostat or Febuxostat group (0.05 g/gCr and - 0.04 g/gCr, respectively). However, the sUA levels decreased significantly in both groups (Topiroxostat group: 8.6 ± 1.1 at baseline to 6.0 ± 1.1 mg/dl at 24 weeks, Febuxostat group: 8.4 ± 1.1 mg/dl at baseline to 5.9 ± 1.3 mg/dl at 24 weeks). No significant change in the eGFR after 24 weeks was noted in either the Topiroxostat or Febuxostat group (- 0.04 ± 4.59 ml/min/1.73 m2 and 0.31 ± 4.70 ml/min/1.73 m2, respectively). CONCLUSIONS In this study, XO inhibitors did not significantly reduce the uPCR in chronic kidney disease stage 3 and 4 patients with hyperuricemia.
Collapse
Affiliation(s)
- Hiroshi Matsuo
- Department of Kidney Center, Suzuka Kaisei Hospital, Suzuka, Japan
| | - Eiji Ishikawa
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan.
| | | | - Yasuhide Mizutani
- Department of Nephrology, Yokkaichi Hazu Medical Center, Yokkaichi, Japan
| | - Akiko Tanoue
- Department of Kidney Center, Suzuka Kaisei Hospital, Suzuka, Japan.,Department of Internal Medicine, Murase Hospital, Suzuka, Japan
| | | | - Tomohiro Murata
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| | - Shinya Okamoto
- Department of Kidney Center, Suzuka Kaisei Hospital, Suzuka, Japan
| | - Toru Ogura
- Clinical Research Support Center, Mie University Hospital, Tsu, Japan
| | - Yuki Nishimura
- Clinical Research Support Center, Mie University Hospital, Tsu, Japan
| | - Hiroo Ito
- Department of Internal Medicine, Nabari City Hospital, Nabari, Japan
| | - Masashi Yasutomi
- Department of Nephrology, Kuwana City Medical Center, Kuwana, Japan
| | - Kan Katayama
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| | - Shinsuke Nomura
- Department of Kidney Center, Suzuka Kaisei Hospital, Suzuka, Japan
| | - Masaaki Ito
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| |
Collapse
|
44
|
Sezai A, Unosawa S, Taoka M, Osaka S, Sekino H, Tanaka M. Changeover Trial of Febuxostat and Topiroxostat for Hyperuricemia with Cardiovascular Disease: Sub-Analysis for Chronic Kidney Disease (TROFEO CKD Trial). Ann Thorac Cardiovasc Surg 2019; 26:202-208. [PMID: 31748427 PMCID: PMC7435131 DOI: 10.5761/atcs.oa.19-00162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background: The TROFEO trial demonstrated that febuxostat causes greater and more rapid reduction of serum uric acid (s-UA) than topiroxostat. We compared these drugs in patients with chronic kidney disease (CKD) by sub-analysis of the TROFEO trial. Methods: This sub-analysis targeted patients with an estimated glomerular filtration rate (eGFR) ≤60 mL/min/1.73 m2. The primary endpoint was the s-UA level. Secondary endpoints included creatinine, eGFR, urinary albumin, cystatin-C, oxidized low-density lipoprotein (Ox-LDL), eicosapentaenoic acid/arachidonic acid ratio, lipid biomarkers, high-sensitivity C-reactive protein, and B-type natriuretic peptide (BNP). Results: There was no significant difference of s-UA between the two groups either before or after treatment. However, s-UA did not exceed 6.0 mg/dL in febuxostat group during the study period, but it exceeded this level in seven patients from topiroxostat group, with the number being significantly higher in topiroxostat group. Serum creatinine (s-Cr) and eGFR were significantly better after 6 months of febuxostat treatment compared with topiroxostat Cystatin-C was significantly lower after 6 months of febuxostat treatment compared with topiroxostat. The Ox-LDL was significantly lower after 3 and 6 months of febuxostat treatment compared with topiroxostat. Conclusion: Febuxostat had stronger renoprotective and antioxidant effects than topiroxostat in patients with hyperuricemia and CKD.
Collapse
Affiliation(s)
- Akira Sezai
- Department of Cardiovascular Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Unosawa
- Department of Cardiovascular Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Makoto Taoka
- Department of Cardiovascular Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Shunji Osaka
- Department of Cardiovascular Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Hisakuni Sekino
- Department of Cardiovascular Surgery, Sekino Hospital, Tokyo, Japan
| | - Masashi Tanaka
- Department of Cardiovascular Surgery, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Furuhashi M, Koyama M, Matsumoto M, Murase T, Nakamura T, Higashiura Y, Tanaka M, Moniwa N, Ohnishi H, Saitoh S, Shimamoto K, Miura T. Annual change in plasma xanthine oxidoreductase activity is associated with changes in liver enzymes and body weight. Endocr J 2019; 66:777-786. [PMID: 31130575 DOI: 10.1507/endocrj.ej19-0053] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Xanthine oxidoreductase (XOR), an enzyme of uric acid formation from hypoxanthine and xanthine, is recognized as a source of oxidative stress. Plasma activity of XOR has been reported to be a biomarker of metabolic disorders associated with obesity, liver dysfunction, insulin resistance, hyperuricemia and adipokines. We investigated longitudinal change in plasma XOR activity, which was determined by using mass spectrometry and liquid chromatography to detect [13C2, 15N2]-uric acid using [13C2, 15N2]-xanthine as a substrate, in 511 subjects (male/female: 244/267) of the Tanno-Sobetsu Study in the years 2016 and 2017. Plasma XOR activity in a basal state was significantly higher in men than in women, but no significant sex difference was observed in annual change in plasma XOR activity. Annual change in plasma activity of XOR was positively correlated with changes in each parameter, including body weight (r = 0.203, p < 0.001), body mass index, diastolic blood pressure, aspartate transaminase (AST) (r = 0.772, p < 0.001), alanine transaminase (r = 0.647, p < 0.001), γ-glutamyl transpeptidase, total cholesterol, triglycerides, uric acid, fasting glucose and HbA1c. Multivariate regression analysis demonstrated that change in AST and that in body weight were independent predictors of change in plasma XOR activity after adjustment of age, sex and changes in each variable with a significant correlation without multicollinearity. In conclusion, annual change in plasma XOR activity is independently associated with changes in liver enzymes and body weight in a general population. Improvement of liver function and reduction of body weight would decrease plasma XOR activity and its related oxidative stress as a therapeutic strategy.
Collapse
Affiliation(s)
- Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masayuki Koyama
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Megumi Matsumoto
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | - Yukimura Higashiura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Norihito Moniwa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shigeyuki Saitoh
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Nursing, Division of Medical and Behavioral Subjects, Sapporo Medical University School of Health Sciences, Sapporo, Japan
| | | | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
46
|
Mizukoshi T, Kato S, Ando M, Sobajima H, Ohashi N, Naruse T, Saka Y, Shimizu H, Nagata T, Maruyama S. Renoprotective effects of topiroxostat for Hyperuricaemic patients with overt diabetic nephropathy study (ETUDE study): A prospective, randomized, multicentre clinical trial. Nephrology (Carlton) 2019; 23:1023-1030. [PMID: 28990729 DOI: 10.1111/nep.13177] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2017] [Indexed: 12/17/2022]
Abstract
AIM We aimed to evaluate the anti-albuminuric effects of topiroxostat in Japanese hyperuricaemic patients with diabetic nephropathy. METHODS In this 24-week, multicentre, open-label, randomized (1 : 1) trial, we assigned hyperuricaemic patients with diabetic nephropathy (estimated glomerular filtration rate ≥ 20 mL/min per 1.73m2 ) and overt proteinuria (0.3 ≤ urine protein to creatinine ratio (UPCR) <3.5 g/g Cr) to either high dose (160 mg daily) or low dose (40 mg daily) topiroxostat. The primary endpoint was the change in albuminuria indicated by urine albumin-to-creatinine ratio (UACR) from the baseline at the final time point. RESULTS A total of 80 patients underwent randomization. The changes in UACR after 24 weeks of treatment (or at the final time point if patients failed to reach 24 weeks) relative to the baseline were -122 mg/gCr (95% CI: -5.1 to -240.1, P = 0.041) in patients treated with high dose, while treatment with low dose topiroxostat could not show significant reduction (P = 0.067). In the linear mixed model including baseline albuminuria, eGFR, age, and sex as covariates, the decreases in UACR were still significant from baseline to 12 weeks by 228.7 ± 83.2 mg/gCr (P = 0.0075) in the high dose group. The adverse-event profile during this study was not different between the groups. CONCLUSION Topiroxostat 160 mg daily reduced albuminuria in patients with diabetic nephropathy. (Funded by Sanwa Kagaku Kenkyusho; Trial registration, UMIN000015403).
Collapse
Affiliation(s)
- Toshihiro Mizukoshi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Sawako Kato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Masahiko Ando
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Aichi, Japan
| | - Hiroshi Sobajima
- Department of Diabetology and Nephrology, Ogaki Municipal Hospital, Aichi, Japan
| | - Norimi Ohashi
- Department of Diabetology and Nephrology, Ogaki Municipal Hospital, Aichi, Japan
| | - Tomohiko Naruse
- Department of Nephrology, Kasugai Municipal Hospital, Aichi, Japan
| | - Yosuke Saka
- Department of Nephrology, Kasugai Municipal Hospital, Aichi, Japan
| | - Hideaki Shimizu
- Department of Nephrology, Chubu Rousai Hospital, Aichi, Japan
| | - Takanobu Nagata
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| |
Collapse
|
47
|
High Serum Uric Acid is Highly Associated with a Reduced Left Ventricular Ejection Fraction Rather than Increased Plasma B-type Natriuretic Peptide in Patients with Cardiovascular Diseases. Sci Rep 2019; 9:682. [PMID: 30679647 PMCID: PMC6346056 DOI: 10.1038/s41598-018-37053-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023] Open
Abstract
High serum uric acid (UA) has been reported to be associated with left ventricular (LV) dysfunction; however, the relationship between UA and plasma B-type natriuretic peptide (BNP), a sensitive biomarker of heart failure, is still unclear. This study investigated their relationship to provide an accurate assessment of high UA. The study patients consisted of 3,077 subjects who underwent cardiac catheterization because of various cardiovascular disorders. Since the explanatory factors of multiple regression analysis were mostly confounding with each other, subgroup analysis was performed by quartering the study population using the respective risk factors and by covariance structure analysis. This analysis revealed that UA was almost always well associated with a reduced LV ejection fraction (LVEF), but generally not with BNP. UA was significantly associated with BNP in lean aged females, but not in obese adolescent males, although LVEF was significantly reduced in response to a high UA in both groups. A high UA is a direct risk factor for cardiac dysfunction from the perspective of BNP; however, augmentation of BNP in response to a high UA would likely be restricted among obese adolescent males. On the other hand, the observed LV systolic dysfunction, such as LVEF, reflects a high UA on an almost constant basis.
Collapse
|
48
|
Pharmacological urate-lowering approaches in chronic kidney disease. Eur J Med Chem 2019; 166:186-196. [PMID: 30769179 DOI: 10.1016/j.ejmech.2019.01.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/20/2019] [Accepted: 01/20/2019] [Indexed: 02/05/2023]
Abstract
Chronic kidney disease (CKD) has become a global public health issue and uric acid (UA) remains a major risk factor of CKD. As the main organ for the elimination of UA, kidney owned a group of urate transporters in tubular epithelium. Kidney disease hampered the UA excretion, and the accumulation of serum UA in return harmed the renal function. Commercially, there are three kinds of agents targeting at urate-lowering, xanthine oxidoreductase inhibitor which prevents the production of UA, uricosuric which increases the concentration of UA in urine thus decreasing serum UA level, and uricase which converts UA to allantoin resulting in the dramatic decrement of serum UA. Of note, in patients with CKD, administration of above-mentioned agents, alone or combined, needs special attention. New evidence is emerging for the efficacy of several urate-lowering drugs for the treatment of hyperuricemia in patients with CKD. Besides, loads of novel and promising drug candidates and phytochemicals are in the different phases of research and development. As of today, there is insufficient evidence to recommend the widespread use of UA-lowering therapy to prevent or slow down the progression of CKD. The review summarized the evidence and perspectives about the treatment of hyperuricemia with CKD for medicinal chemist and nephrologist.
Collapse
|
49
|
Inoue MK, Yamamotoya T, Nakatsu Y, Ueda K, Inoue Y, Matsunaga Y, Sakoda H, Fujishiro M, Ono H, Morii K, Sasaki K, Masaki T, Suzuki Y, Asano T, Kushiyama A. The Xanthine Oxidase Inhibitor Febuxostat Suppresses the Progression of IgA Nephropathy, Possibly via Its Anti-Inflammatory and Anti-Fibrotic Effects in the gddY Mouse Model. Int J Mol Sci 2018; 19:E3967. [PMID: 30544662 PMCID: PMC6320819 DOI: 10.3390/ijms19123967] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/28/2018] [Accepted: 12/05/2018] [Indexed: 12/29/2022] Open
Abstract
Recent clinical studies have demonstrated the protective effect of xanthine oxidase (XO) inhibitors against chronic kidney diseases, although the underlying molecular mechanisms remain unclear. However, to date, neither clinical nor basic research has been carried out to elucidate the efficacy of XO inhibitor administration for IgA nephropathy. We thus investigated whether febuxostat, an XO inhibitor, exerts a protective effect against the development of IgA nephropathy, using gddY mice as an IgA nephropathy rodent model. Eight-week-old gddY mice were provided drinking water with (15 μg/mL) or without febuxostat for nine weeks and then subjected to experimentation. Elevated serum creatinine and degrees of glomerular sclerosis and fibrosis, judged by microscopic observations, were significantly milder in the febuxostat-treated than in the untreated gddY mice, while body weights and serum IgA concentrations did not differ between the two groups. In addition, elevated mRNA levels of inflammatory cytokines such as TNFα, MCP-1, IL-1β, and IL-6, collagen isoforms and chemokines in the gddY mouse kidneys were clearly normalized by the administration of febuxostat. These data suggest a protective effect of XO inhibitors against the development of IgA nephropathy, possibly via suppression of inflammation and its resultant fibrotic changes, without affecting the serum IgA concentration.
Collapse
Affiliation(s)
- Masa-Ki Inoue
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Takeshi Yamamotoya
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Yusuke Nakatsu
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Koji Ueda
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Yuki Inoue
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Yasuka Matsunaga
- Center for Translational Research in Infection & Inflammation, School of Medicine, Tulane University, 6823 St. Charles Avenue, New Orleans, LA 70118, USA.
| | - Hideyuki Sakoda
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | - Midori Fujishiro
- Division of Diabetes and Metabolic Diseases, Nihon University School of Medicine, Itabashi, Tokyo 173-8610, Japan.
| | - Hiraku Ono
- Department of Clinical Cell Biology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba 260-8670, Japan.
| | - Kenichi Morii
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Yusuke Suzuki
- Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Tomoichiro Asano
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-8551, Japan.
| | - Akifumi Kushiyama
- Division of Diabetes and Metabolism, The Institute for Adult Diseases, Asahi Life Foundation, Chuo-ku, Tokyo 103-0002, Japan.
| |
Collapse
|
50
|
Furuhashi M, Matsumoto M, Tanaka M, Moniwa N, Murase T, Nakamura T, Ohnishi H, Saitoh S, Shimamoto K, Miura T. Plasma Xanthine Oxidoreductase Activity as a Novel Biomarker of Metabolic Disorders in a General Population. Circ J 2018; 82:1892-1899. [PMID: 29643318 DOI: 10.1253/circj.cj-18-0082] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
BACKGROUND Xanthine oxidoreductase (XOR) is an enzyme that catalyzes the formation of uric acid from hypoxanthine and xanthine, leading to an increase in superoxide and reactive oxygen species. Activation of XOR promotes oxidative stress-related tissue injury. We investigated the associations between metabolic parameters and plasma XOR activity measured by a sensitive and accurate assay using a combination of liquid chromatography and triple quadrupole mass spectrometry to detect [13C2,15N2]-uric acid using [13C2,15N2]-xanthine as a substrate. METHODS AND RESULTS A total of 627 Japanese subjects (M/F, 292/335) from the Tanno-Sobetsu Study, a population-based cohort, were recruited. Plasma XOR activity was significantly higher in males than in females, and habitual smoking was associated with elevation of activity. Plasma XOR activity was positively correlated with body mass index (BMI; r=0.323, P<0.001), waist circumference, blood pressure, and levels of liver enzymes including alanine transaminase (r=0.694, P<0.001), uric acid (r=0.249, P<0.001), triglycerides (r=0.312, P<0.001), hemoglobin A1c, fasting glucose, insulin and HOMA-R (r=0.238, P<0.001) as a marker of insulin resistance and was negatively correlated with high-density lipoprotein cholesterol level. On stepwise and multivariate regression analyses, BMI, smoking and levels of alanine transaminase, uric acid, triglycerides and HOMA-R were independent predictors of plasma XOR activity after adjustment for age and gender. CONCLUSIONS Plasma XOR activity is a novel biomarker of metabolic disorders in a general population.
Collapse
Affiliation(s)
- Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| | - Megumi Matsumoto
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| | - Norihito Moniwa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| | | | | | - Hirofumi Ohnishi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
- Department of Public Health, Sapporo Medical University School of Medicine
| | - Shigeyuki Saitoh
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
- Department of Nursing, Division of Medical and Behavioral Subjects, Sapporo Medical University School of Health Sciences
| | | | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine
| |
Collapse
|