1
|
Yang T, Luo L, Luo X, Liu X. Metabolic crosstalk and therapeutic interplay between diabetes and hyperuricemia. Diabetes Res Clin Pract 2025; 224:112204. [PMID: 40294652 DOI: 10.1016/j.diabres.2025.112204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
Hyperuricemia and diabetes mellitus (DM) are prevalent metabolic disorders with high comorbidity, imposing a substantial global public health burden. Their coexistence is not merely additive but synergistic, exacerbating metabolic dysregulation through mechanisms such as insulin resistance and β-cell apoptosis, ultimately establishing a vicious cycle. Both disorders induce acute and chronic damage to vital organs, particularly the cardiovascular, renal systems. Hyperuricemia aggravates diabetic complications, notably diabetic cardiomyopathy, nephropathy and retinopathy via oxidative stress, inflammation, and metabolic dysregulation.Current urate-lowering therapies (ULTs), such as xanthine oxidase inhibitors and urate transporter 1 (URAT1, also known as SLC22A12) antagonists, demonstrate potential benefits in ameliorating diabetic complications but face challenges including safety concerns and dose adjustments. Similarly, several glucose-lowering drugs also exhibit the benefits of improving hyperuricemia. This review summarizes the metabolic crosstalk and therapeutic interplay between hyperuricemia and DM, examines the pathogenic role of uric acid in diabetic complications, and discusses the benefits and challenges of existing ULTs and glucose-lowering drugs in disrupting this cycle of metabolic dysregulation and concurrent organ damage. We hope our findings deepen the comprehension of the intricate metabolic crosstalk between glucose and urate homeostasis, providing novel therapeutic insights for patients with comorbid DM and hyperuricemia.
Collapse
Affiliation(s)
- Tianshu Yang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China
| | - Lingyun Luo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030 Hubei, China
| | - Xuelian Luo
- Department of Oncology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Xiaolei Liu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 Hubei, China; Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan 430030 Hubei, China.
| |
Collapse
|
2
|
Timsans J, Kauppi J, Rantalaiho V, Kerola A, Hakkarainen K, Lehto T, Kautiainen H, Kauppi M. Serum Uric Acid Is Associated with Insulin Resistance in Non-Diabetic Subjects. J Clin Med 2025; 14:2621. [PMID: 40283451 PMCID: PMC12028212 DOI: 10.3390/jcm14082621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Glucose metabolism disorders are major contributors to morbidity and mortality. Elevated serum uric acid (SUA) is closely linked to the cardiometabolic consequences of glucose metabolism disorders, various other comorbidities, and mortality. In this study, we explore the relationship between SUA and fasting plasma glucose (FPG), insulin levels, and insulin resistance in an older Finnish adult cohort. Methods: We used data from the GOAL (GOod Ageing in Lahti region) study-a prospective, population-based study of Finnish individuals aged 52-76 years. A total of 2322 non-diabetic subjects were included in the study. Data of SUA, FPG, and other laboratory parameters, comorbidities, lifestyle habits, and socioeconomic factors were collected. Subjects with SUA values of >410 μmol/L (≈6.9 mg/dL; 75th percentile) were regarded as hyperuricemic. We investigated the relationship between SUA and FPG, insulin levels, and insulin resistance [homeostatic model assessment of insulin resistance (HOMA-IR) ≥2.65]. Results: We found statistically significant sex-, age- and BMI-adjusted small to moderate relationships (Cohen's standard for β values above 0.10 and 0.30, respectively) between SUA and FPG, insulin levels, and insulin resistance in the whole study population as well as in the female and male subgroups. The higher the SUA level, the higher the HOMA-IR [(adjusted β = 0.21 (95% CI: 0.17 to 0.25)], and it rises drastically if SUA is above 400 μmol/L (≈6.7 mg/dL). The probability of a subject having insulin resistance is related to SUA level. Conclusions: Hyperuricemia is associated with elevated FPG and insulin resistance, emphasizing the importance of addressing both conditions. Further research may explore hyperuricemia treatment's role in preventing glucose metabolism disorders and their cardiometabolic consequences.
Collapse
Affiliation(s)
- Janis Timsans
- Department of Rheumatology, Päijät-Häme Central Hospital, Wellbeing Services County of Päijät-Häme, 15850 Lahti, Finland; (A.K.); (M.K.)
- Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland;
| | - Jenni Kauppi
- Unit of Physiatry and Rehabilitation Medicine, Päijät-Häme Central Hospital, Wellbeing Services County of Päijät-Häme, 15850 Lahti, Finland;
| | - Vappu Rantalaiho
- Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland;
- Centre for Rheumatic Diseases, Tampere University Hospital, 33521 Tampere, Finland
- Department of Medicine, Kanta-Häme Central Hospital, 13530 Hämeenlinna, Finland
| | - Anne Kerola
- Department of Rheumatology, Päijät-Häme Central Hospital, Wellbeing Services County of Päijät-Häme, 15850 Lahti, Finland; (A.K.); (M.K.)
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Kia Hakkarainen
- Department of Nephrology, Päijät-Häme Central Hospital, Wellbeing Services County of Päijät-Häme, 15850 Lahti, Finland;
| | - Tiina Lehto
- Department of Clinical Chemistry, Fimlab Laboratoriot Oy, 15140 Lahti, Finland;
| | - Hannu Kautiainen
- Folkhälsan Research Center, 00250 Helsinki, Finland;
- Primary Health Care Unit, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Markku Kauppi
- Department of Rheumatology, Päijät-Häme Central Hospital, Wellbeing Services County of Päijät-Häme, 15850 Lahti, Finland; (A.K.); (M.K.)
- Clinicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
3
|
Gu W, Zhao J, Xu Y. Hyperuricemia-induced complications: dysfunctional macrophages serve as a potential bridge. Front Immunol 2025; 16:1512093. [PMID: 39935474 PMCID: PMC11810932 DOI: 10.3389/fimmu.2025.1512093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
With the changes in modern life, hyperuricemia (HUA) has become a serious universal health issue, leading to rising morbidity and mortality. Characterized by elevated levels of UA, HUA has become an independent risk factor for gout, chronic kidney disease, insulin resistance, cardiovascular disease, nonalcoholic fatty liver disease, etc. As HUA is a metabolic syndrome, the immune response is likely to play an active role throughout the whole process. Moreover, macrophages, as an indispensable component of the immune system, may serve as a promising target for addressing hyperuricemia-induced inflammation. Along with their precursor cells, monocytes, macrophages play a key role in the pathogenesis of HUA, primarily through three specific aspects, all of which are associated with inflammatory cytokines. The first mechanism involves direct action on urate transporters, such as URAT1 and ABCG2. The second mechanism is the modulation of inflammation, including targeting toll-like receptors (TLRs) and the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome. The third mechanism pertains to the effects on oxidative stress mediators. In this review, we summarize the underlying mechanisms of hyperuricemia, focusing on the effects of macrophages, therapeutic approaches, and clinical trials addressing hyperuricemia-caused dysfunction. Additionally, we highlight directions for future development, aiming to support future theoretical studies.
Collapse
Affiliation(s)
- Wenyi Gu
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiajing Zhao
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Yu Xu
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for Traditional Chinese Medicine New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Yang XH, Zhang BL, Cheng Y, Fu SK, Jin HM. Febuxostat provides renoprotection in patients with hyperuricemia or gout: a systematic review and meta-analysis of randomized controlled trials. Ann Med 2024; 56:2332956. [PMID: 38738384 PMCID: PMC11095284 DOI: 10.1080/07853890.2024.2332956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 03/09/2024] [Indexed: 05/14/2024] Open
Abstract
PURPOSE It is unknown whether febuxostat can delay the progression of kidney dysfunction and reduce kidney endpoint events. The aim was to evaluate the renoprotective effect of febuxostat in patients with hyperuricemia or gout by performing a meta-analysis of randomized controlled trials (RCTs). METHODS MEDLINE, Web of science, EMBASE, ClinicalTrials.gov, and the Cochrane Central Register for Randomized Controlled Trials were searched. The main outcomes included kidney events (serum creatinine doubling or progression to end-stage kidney disease or dialysis). The secondary outcomes were the rate of change in the estimated glomerular filtration rate (eGFR) and changes in the urine protein or urine albumin to creatinine ratio from baseline to the end of follow-up. We used random-effects models to calculate the pooled risk estimates and 95% CIs. RESULTS A total of 16 RCTs were included in the meta-analysis. In comparison with the control group, the patients who received febuxostat showed a reduced risk of kidney events (RR = 0.56, 95% CI 0.37-0.84, p = 0.006) and a slower decline in eGFR (WMD = 0.90 mL/min/1.73 m2, 95% CI 0.31-1.48, p = 0.003). The pooled results also revealed that febuxostat use reduced the urine albumin to creatinine ratio (SMD = -0.21, 95% CI -0.41 to -0.01, p = 0.042). CONCLUSION Febuxostat use is associated with a reduced risk of kidney events and a slow decline in eGFR. In addition, the urine albumin to creatinine ratio decreased in febuxostat users. Accordingly, it is an effective drug for delaying the progression of kidney function deterioration in patients with gout.Systematic review registration: PROSPERO CRD42021272591.
Collapse
Affiliation(s)
- Xiu Hong Yang
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, China
| | - Bao Long Zhang
- The Institutes of Biomedical Sciences (IBS), Fudan University, Shanghai, China
| | - Yun Cheng
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China
| | - Shun Kun Fu
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China
| | - Hui Min Jin
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China
- Department of Nephrology, The People’s Hospital of Wenshan Prefecture, Yunnan Province, China
| |
Collapse
|
5
|
El-Shoura EAM, Sharkawi SMZ, Abdelzaher LA, Abdel-Wahab BA, Ahmed YH, Abdel-Sattar AR. Reno-protective effect of fenofibrate and febuxostat against vancomycin-induced acute renal injury in rats: Targeting PPARγ/NF-κB/COX-II and AMPK/Nrf2/HO-1 signaling pathways. Immunopharmacol Immunotoxicol 2024; 46:509-520. [PMID: 38918173 DOI: 10.1080/08923973.2024.2373216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Vancomycin (VCM) is used clinically to treat serious infections caused by multi-resistant Gram-positive bacteria, although its use is severely constrained by nephrotoxicity. This study investigated the possible nephroprotective effect of febuxostat (FX) and/or fenofibrate (FENO) and their possible underlying mechanisms against VCM-induced nephrotoxicity in a rat model. METHODS Male Wistar rats were randomly allocated into five groups; Control, VCM, FX, FENO, and combination groups. Nephrotoxicity was evaluated histopathologically and biochemically. The oxidative stress biomarkers (SOD, MDA, GSH, total nitrite, GPx, MPO), the apoptotic marker, renal Bcl-2 associated X protein (Bax), and inflammatory and kidney injury markers (IL-1β, IL-6, TNF-α, Nrf2, OH-1, kappa-light-chain-enhancer of activated B cells (NF-κB), NADPH oxidase, Kim-1, COX-II, NGAL, Cys-C were also evaluated. RESULTS VCM resulted in significant elevation in markers of kidney damage, oxidative stress, apoptosis, and inflammatory markers. Co-administration of VCM with either/or FX and FENO significantly mitigated nephrotoxicity and associated oxidative stress, inflammatory and apoptotic markers. In comparison to either treatment alone, a more notable improvement was observed with the FX and FENO combination regimen. CONCLUSION Our findings show that FX, FENO, and their combination regimen have a nephroprotective impact on VCM-induced kidney injury by suppressing oxidative stress, apoptosis, and the inflammatory response. Renal recovery from VCM-induced injury was accomplished by activation of Nrf2/HO-1 signaling and inhibition of NF-κB expression. This study highlights the importance of FX and FENO as effective therapies for reducing nephrotoxicity in VCM-treated patients.
Collapse
Affiliation(s)
- Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
- Department of Pharmacy Practice, Faculty of Pharmacy, Horus University in Egypt, New Damietta, Egypt
| | - Souty M Z Sharkawi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Lobna A Abdelzaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Basel A Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Yasmine H Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | |
Collapse
|
6
|
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
|
7
|
Takeshita M, Tanaka A, Yoshida H, Nakamura I, Shibata Y, Hata S, Kushiyama A, Okutsu M, Ishizu T, Node K. Effect of the Xanthine Oxidase Inhibitor, Febuxostat, on WBC Count in Asymptomatic Hyperuricemia: Subanalysis of the Randomized PRIZE Study. J Atheroscler Thromb 2024; 31:864-875. [PMID: 38220209 PMCID: PMC11150717 DOI: 10.5551/jat.64574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/19/2023] [Indexed: 01/16/2024] Open
Abstract
AIMS The anti-inflammatory effects of the xanthine oxidase inhibitor, febuxostat, a urate-lowering agent, have been reported in animal studies. However, the anti-inflammatory effects of urate-lowering therapy and its associated cardiovascular protective effects have not been fully determined in actual clinical practice. This study aimed to investigate the effect of febuxostat on white blood cell (WBC) count in patients with asymptomatic hyperuricemia and to assess for potential correlations between changes in WBC count and inflammatory biomarkers and atherosclerosis in this patient population. METHODS This was a post hoc subanalysis of the PRIZE study, a multicenter, prospective, randomized, open-label clinical trial. In the PRIZE study, asymptomatic hyperuricemia patients were randomized to febuxostat group or control group with non-pharmacological therapy and evaluated the effect on vascular. The primary endpoints of this study were the assessment of the time course of WBC count over 24 months and its changes from baseline. Correlations of WBC count with high-sensitivity C-reactive protein (hs-CRP) and mean common carotid artery (CCA)-IMT were also exploratorily examined in the febuxostat group. RESULTS A total of 444 patients (febuxostat group, n=223; control group, n=221) with WBC measurements available at baseline and at least one of the follow-up time points of 12 or 24 months, were enrolled. Febuxostat modestly, but significantly, reduced WBC counts at 12 and 24 months compared with the baseline levels (P=0.002 and P=0.026, respectively). Notably, the WBC count in the febuxostat group at 12 and 24 months was significantly lower than that in the control group (P=0.007 and P=0.023, respectively). The changes in WBC count were associated with those of hs-CRP (P=0.038), but not with CCA-IMT (P=0.727). CONCLUSIONS Febuxostat therapy for 24 months modestly, but significantly, decreased WBC count in patients with asymptomatic hyperuricemia. This might potentially reflect a modest anti-inflammatory action of febuxostat in clinical settings.
Collapse
Affiliation(s)
| | - Atsushi Tanaka
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Hisako Yoshida
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Ikuko Nakamura
- Department of Cardiovascular Medicine, Saga-Ken Medical Centre Koseikan, Saga, Japan
| | - Yoshisato Shibata
- Department of Cardiology, Miyazaki Medical Association Hospital, Miyazaki, Japan
| | - Shiro Hata
- Department of Cardiology, Sasebo City General Hospital, Nagasaki, Japan
| | - Akifumi Kushiyama
- Department of Pharmacotherapy, Meiji Pharmaceutical University, Tokyo, Japan
| | - Masaaki Okutsu
- Department of Cardiovascular Medicine, New Tokyo Hospital, Matsudo, Japan
| | - Tomoko Ishizu
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| |
Collapse
|
8
|
Wang Y, Chang J, Qiao S, Yang Y, Yun C, Li Y, Wang F. Salvianolic acid B attenuates diabetic nephropathy through alleviating ADORA2B, NALP3 in flammasome, and NFκB activity. Can J Physiol Pharmacol 2024; 102:318-330. [PMID: 38070193 DOI: 10.1139/cjpp-2023-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Diabetic nephropathy is one of the microvascular complications of diabetes. This study is aimed at investigating the role and mechanisms of salvianolic acid B (Sal B) in diabetic nephropathy. High glucose (HG)-induced human renal tubular epithelial HK-2 cells were treated with Sal B, BAY-60-6583 (agonist of adenosine 2B receptor), or PSB-603 (antagonist of adenosine 2B receptor) for 24 h. Adenosine A2b receptor (ADORA2B), NACHT, leucine-rich repeat (LRR), and pyrin (PYD) domains-containing protein 3 (NALP3), and nuclear factor Kappa B (NFκB) expressions, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) levels were examined. Following 6 weeks of Sal B treatment, db/db mice blood and kidney tissue were harvested for biochemical detection with hematoxylin-eosin (H&E), Masson's, periodic acid schiff (PAS), and Sirius red staining and detection of ADORA2B, NALP3, NFκB, interleukin 1β (IL-1β), and toll-like receptor 4 (TLR4) activity. NFκB, NALP3, and ADORA2B were found to be downregulated in Sal B treated HK-2 cells exposed to high glucose (HG), accompanied by elevated levels of MMPs and reduced intracellular ROS production. Sal B-treated diabetic mice had the improvement in body weight, water intake, hyperglycemia, hyperlipidemia, and liver and kidney function. Altogether, Sal B attenuates HG-induced kidney tubule cell injury and diabetic nephropathy in diabetic mice, providing clues to other novel mechanisms by which Sal B is beneficial in diabetic nephropathy.
Collapse
Affiliation(s)
- Ying Wang
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, Hainan, China
| | - Jiang Chang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, Hainan, China
| | - Shubin Qiao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, Hainan, China
- Department of Pulmonary and Critical Care Medicine, Beijing 100071, China
| | - Ying Yang
- Department of Endocrinology, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China
| | - Chuan Yun
- Department of Endocrinology, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, Hainan, China
| | - Yongyan Li
- Department of Nephrology, Hainan Medical University, Haikou 570102, Hainan, China
| | - Fa Wang
- Department of Anesthesiology, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
9
|
Bai Y, Shi H, Zhang Y, Zhang C, Wu B, Wu X, Fang Z, Wang Q, Sima X, Zhang T. Febuxostat attenuates secondary brain injury caused by cerebral hemorrhage through inhibiting inflammatory pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:740-746. [PMID: 38645501 PMCID: PMC11024405 DOI: 10.22038/ijbms.2024.74655.16212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/27/2023] [Indexed: 04/23/2024]
Abstract
Objectives Neuroinflammation is considered an important step in the progression of secondary brain injury (SBI) induced by cerebral hemorrhage (ICH). The nucleotide-binding and oligomerization structural domain-like receptor family of pyridine structural domain-containing 3 (NLRP3) inflammasomes play an important role in the immune pathophysiology of SBI. Febuxostat (Feb) is a xanthine oxidase inhibitor that is approved for the treatment of gout and has been found to have potent anti-inflammatory effects. However, it has been less studied after ICH and we aimed to explore its protective role in ICH. Materials and Methods We established an autologous blood-brain hemorrhage model in C57BL/6 mice. Functions of co-expressed genes were analyzed by trend analysis and bioinformatics analysis. Enzyme-linked immunosorbent assay were used to assess the inflammatory factor levels. Fluoro-Jade B histochemistry and TUNEL staining were used to detect neuronal apoptosis. Immunofluorescence staining and western blotting were used to detect the expression of NLRP3 inflammasomes. Results Pretreatment with Feb reduced neuronal cell death and degeneration and alleviated neurobehavioral disorders in vivo. Feb was found to modulate inflammation-related pathways by trend analysis and bioinformatics analysis. In addition, Feb inhibited microglia activation and elevated cytokine levels after ICH. Furthermore, double immunofluorescence staining showed that co-localization of NLRP3 with Iba1 positive cells was reduced after treatment with Feb. Finally, we found that Feb inhibited the activation of the NLRP3/ASC/caspase-1 pathway after ICH. Conclusion By inhibiting the NLRP3 inflammasome, preconditioning Feb attenuates inflammatory injury after ICH. Our findings may provide new insights into the role of Feb in neuroprotection.
Collapse
Affiliation(s)
- Yang Bai
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- These authors contributed equally to this work
| | - Hongxia Shi
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- These authors contributed equally to this work
| | - Ying Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenyu Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Bin Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xinghan Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenwei Fang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qi Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiutian Sima
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tiejun Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Wang Y, Lu J. The Management of Diabetes with Hyperuricemia: Can We Hit Two Birds with One Stone? J Inflamm Res 2023; 16:6431-6441. [PMID: 38161355 PMCID: PMC10757772 DOI: 10.2147/jir.s433438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 01/03/2024] Open
Abstract
Serum urate (SU) is an independent predictor for the incidence of diabetes. In current diabetes treatment regimens, there is insufficient appreciation of the importance of hyperuricemia (HU) in disease control and prevention. To summarize the updated knowledge on the effects of SU on β-cell function, insulin resistance and chronic diabetic complications, as well as to evaluate the management of patients with both HU and diabetes, we searched the MEDLINE PubMed database, and included 285 journal articles. An inverted U-shaped relationship between fasting plasma glucose and SU levels was established in this review. Elevated SU levels may enhance the development of chronic diabetic complications, including macrovascular and microvascular dysfunction. Diet and exercise are essential parts of the lifestyle changes necessary for HU and diabetes management. Glucose- and urate-lowering drug selection and combination should be made with the principle of ameliorating, and at least not deteriorating, diabetes and HU. Medical artificial intelligence technology and monitoring systems can help to improve the effectiveness of long-term management of HU and diabetes through digital healthcare. This study comprehensively reviews and provides a scientific and reliable basis for and viewpoints on the clinical management of diabetes and HU.
Collapse
Affiliation(s)
- Yunyang Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jie Lu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
- Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
11
|
Teragawa H, Tanaka A, Fujii Y, Yoshida H, Ueda T, Nomura S, Kadokami T, Koide H, Saito M, Sano H, Bando YK, Murohara T, Node K, for the PRIZE Study Investigators. Effect of febuxostat on the level of malondialdehyde-modified low-density lipoprotein, an oxidative stress marker: A subanalysis of the PRIZE study. Clin Cardiol 2023; 46:698-706. [PMID: 36991567 PMCID: PMC10270262 DOI: 10.1002/clc.24014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Febuxostat is a selective xanthine oxidase inhibitor that reportedly exhibits antioxidant properties. We previously performed a multicentre, randomized controlled (PRIZE) study for vascular evaluation under uric acid (UA) control by febuxostat to investigate the progression of carotid lesions in asymptomatic hyperuricemic patients with carotid atherosclerosis for 2 years. HYPOTHESIS The current subanalysis of the PRIZE study aimed to assess the effect of febuxostat on the level of malondialdehyde-modified low-density lipoprotein (MDA-LDL), an oxidative stress marker. METHODS We recruited 383 patients (febuxostat group, n = 200; control group, n = 183) from the PRIZE trial for whom MDA-LDL measurements were available. The UA, MDA-LDL, low-density lipoprotein cholesterol (LDL-C) levels, and MDA-LDL/LDL-C ratio were identified, represented as the estimated difference from baseline to 24 months. We also evaluated the relationship between febuxostat dose (10, ≤20 to <40, and ≤40 to ≤60 mg) and changes in the MDA-LDL level, LDL-C level, or MDA-LDL/LDL-C ratios. RESULTS The estimated change in MDA-LDL/LDL-C ratio from baseline to 24 months was significantly lower in the febuxostat group than in the control group (p = .025), whereas the estimated changes in MDA-LDL (p = .235) and LDL-C (p = .323) levels did not differ between the two groups. No significant correlation existed between the febuxostat doses and the estimated change in the MDA-LDL level (p = .626), LDL-C level (p = .896), or MDA-LDL/LDL-C ratio (p = .747). CONCLUSIONS Our findings may indicate a possibility that febuxostat can lower the MDA-LDL/LDL-C ratio, a potential marker of atherosclerosis and oxidative stress, in asymptomatic hyperuricemic patients with carotid atherosclerosis. Further studies are required to validate our findings and elucidate the clinical antioxidant effect of febuxostat.
Collapse
Affiliation(s)
- Hiroki Teragawa
- Department of Cardiovascular MedicineJR Hiroshima HospitalHiroshimaJapan
| | - Atsushi Tanaka
- Department of Cardiovascular MedicineSaga UniversitySagaJapan
| | - Yuichi Fujii
- Department of Cardiovascular MedicineJR Hiroshima HospitalHiroshimaJapan
| | - Hisako Yoshida
- Department of Medical StatisticsOsaka Metropolitan University Graduate School of MedicineOsakaJapan
| | - Tomohiro Ueda
- Department of Cardiovascular MedicineJR Hiroshima HospitalHiroshimaJapan
| | - Shuichi Nomura
- Department of Cardiovascular MedicineJR Hiroshima HospitalHiroshimaJapan
| | - Toshiaki Kadokami
- Department of Cardiovascular MedicineSaiseikai Futsukaichi HospitalFukuokaJapan
| | - Hisashi Koide
- Department of Diabetes, Metabolism and EndocrinologyChiba University HospitalChibaJapan
| | - Makoto Saito
- Department of Internal MedicineNishio Municipal HospitalNishioAichiJapan
| | - Hiroaki Sano
- Department of CardiologyNagoya Ekisaikai HospitalNogayaAichiJapan
| | - Yasuko K. Bando
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Toyoaki Murohara
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Koichi Node
- Department of Cardiovascular MedicineSaga UniversitySagaJapan
| | | |
Collapse
|
12
|
Kitamura Y, Oikawa S, Chang J, Mori Y, Ichihara G, Ichihara S. Carbonylated Proteins as Key Regulators in the Progression of Metabolic Syndrome. Antioxidants (Basel) 2023; 12:antiox12040844. [PMID: 37107219 PMCID: PMC10135001 DOI: 10.3390/antiox12040844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Based on the known role of oxidative stress in the pathogenesis and progression of metabolic syndrome, we used two-dimensional gel electrophoresis with immunochemical detection of protein carbonyls (2D-Oxyblot) to characterize the carbonylated proteins induced by oxidative stress in spontaneously hypertensive rats/NDmcr-cp (CP), an animal model of metabolic syndrome. We also profiled the proteins that showed change of expression levels in their epididymal adipose tissue at the pre-symptomatic (6-week-old) and the symptomatic (25-week-old) stages of the metabolic syndrome. Two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) combined with matrix-assisted laser desorption ionization time-of-flight tandem mass spectrometry (MALDI-TOF/TOF MS) was used to analyze proteins extracted from the epididymal adipose tissue. The up-regulated proteins identified at the pre-symptomatic stage were mainly associated with ATP production and redox reaction, while the down-regulated proteins found at the symptomatic stage were involved in antioxidant activity and the tricarboxylic acid (TCA) cycle. Further analysis using the 2D-Oxyblot showed significantly high carbonylation levels of gelsolin and glycerol-3-phosphate dehydrogenase [NAD+] at the symptomatic stage. These results suggest that reduced antioxidant capacity underlies the increased oxidative stress state in the metabolic syndrome. The identified carbonylated proteins, including gelsolin, are potential targets that may act as key regulators in the progression of the metabolic syndrome.
Collapse
Affiliation(s)
- Yuki Kitamura
- Department of Molecular and Environmental Medicine, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke 329-0498, Japan
| | - Shinji Oikawa
- Department of Molecular and Environmental Medicine, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Jie Chang
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan
| | - Yurie Mori
- Department of Molecular and Environmental Medicine, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Tokyo University of Sciences, Noda 278-8510, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke 329-0498, Japan
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan
| |
Collapse
|
13
|
Zhu L, Wang X, Sun J, Qian Q, Yu J, An X. Hyperuricemia Predicts the Progression of Type 2 Diabetic Kidney Disease in Chinese Patients. Diabetes Ther 2023; 14:581-591. [PMID: 36757669 PMCID: PMC9981872 DOI: 10.1007/s13300-023-01374-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
INTRODUCTION Diabetic kidney disease (DKD) has a high global disease burden and substantially increases the risk of end-stage renal disease and cardiovascular events. High levels of serum uric acid (SUA), or hyperuricemia, may indicate patients with type 2 diabetes (T2D) at risk for kidney disease. METHODS This study explored the association between SUA levels and progression of kidney disease among patients with T2D. A cross-sectional study of 993 Chinese patients aged 20-75 years with T2D and DKD was conducted. Patients were stratified by progression risk of kidney disease based on estimated glomerular filtration rate and ratio of urinary albumin to creatinine, according to Kidney Disease: Improving Global Outcomes (KDIGO) criteria. Ordinal logistic regression was used to assess associations between SUA and different KDIGO risk categories. RESULTS Among 768 patients in the final analysis, those with hyperuricemia and higher SUA were more likely to be assigned to higher KDIGO risk categories. Patients with SUA > 420 μmol/L were ninefold more likely to be in a higher KDIGO risk category than those with SUA < 300 μmol/L (odds risk 9.74, 95% confidence interval 5.47-17.33, P < 0.001). CONCLUSIONS Hyperuricemia may be associated with higher risk of DKD progression in individuals with T2D.
Collapse
Affiliation(s)
- Lin Zhu
- Physical Examination Center, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Xuening Wang
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jiaxing Sun
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Qi Qian
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jiangyi Yu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| | - Xiaofei An
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
14
|
Yamamoto K, Yamashita M, Oda M, Tjendana Tjhin V, Inagawa H, Soma GI. Oral Administration of Lipopolysaccharide Enhances Insulin Signaling-Related Factors in the KK/Ay Mouse Model of Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24054619. [PMID: 36902049 PMCID: PMC10003108 DOI: 10.3390/ijms24054619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
Lipopolysaccharide (LPS), an endotoxin, induces systemic inflammation by injection and is thought to be a causative agent of chronic inflammatory diseases, including type 2 diabetes mellitus (T2DM). However, our previous studies found that oral LPS administration does not exacerbate T2DM conditions in KK/Ay mice, which is the opposite of the response from LPS injection. Therefore, this study aims to confirm that oral LPS administration does not aggravate T2DM and to investigate the possible mechanisms. In this study, KK/Ay mice with T2DM were orally administered LPS (1 mg/kg BW/day) for 8 weeks, and blood glucose parameters before and after oral administration were compared. Abnormal glucose tolerance, insulin resistance progression, and progression of T2DM symptoms were suppressed by oral LPS administration. Furthermore, the expressions of factors involved in insulin signaling, such as insulin receptor, insulin receptor substrate 1, thymoma viral proto-oncogene, and glucose transporter type 4, were upregulated in the adipose tissues of KK/Ay mice, where this effect was observed. For the first time, oral LPS administration induces the expression of adiponectin in adipose tissues, which is involved in the increased expression of these molecules. Briefly, oral LPS administration may prevent T2DM by inducing an increase in the expressions of insulin signaling-related factors based on adiponectin production in adipose tissues.
Collapse
Affiliation(s)
- Kazushi Yamamoto
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
| | - Masashi Yamashita
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
| | - Masataka Oda
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
| | - Vindy Tjendana Tjhin
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
| | - Hiroyuki Inagawa
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
- Research Institute for Healthy Living, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-0841, Japan
| | - Gen-Ichiro Soma
- Control of Innate Immunity, Technology Research Association, Takamatsu 761-0301, Japan
- Research Institute for Healthy Living, Niigata University of Pharmacy and Applied Life Sciences, Niigata 956-0841, Japan
- Correspondence: ; Tel.: +81-87-813-9201
| |
Collapse
|
15
|
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
|
16
|
Xu C, Meng J. Febuxostat, a potential drug in the secondary prevention of cardiovascular disease with hyperuricemia. Int J Cardiol 2023; 370:367. [PMID: 36306952 DOI: 10.1016/j.ijcard.2022.10.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Can Xu
- The First Affiliated Hospital of University of South China, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Jun Meng
- The First Affiliated Hospital of University of South China, Department of Function, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
17
|
Febuxostat Protects Human Aortic Valve Endothelial Cells From Oxidized Low-density Lipoprotein-Induced Injury and Monocyte Attachment. J Cardiovasc Pharmacol 2022; 80:861-868. [PMID: 35881896 DOI: 10.1097/fjc.0000000000001326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 06/14/2022] [Indexed: 12/13/2022]
Abstract
ABSTRACT Atherosclerosis (AS) is a common cardiovascular disease with high morbidity and mortality. The pathogenesis of AS is closely related to endothelial dysfunction, which is mainly induced by oxidative stress, inflammation, and enhanced adhesion of monocytes to endothelial cells on the vessel wall. Febuxostat is a novel antigout agent recently reported to exert protective effects on endothelial dysfunction. This study aims to investigate the protective capacity of febuxostat against oxidized low-density lipoprotein (ox-LDL)-induced injury and monocyte attachment to endothelial cells. Human aortic valve endothelial cells (HAVECs) were stimulated with ox-LDL in the presence or absence of febuxostat (5 and 10 μM) for 6 hours. Mitochondrial reactive oxygen species were measured using MitoSox red staining, and the level of protein carbonyl was detected using enzyme-linked immunosorbent assay (ELISA). The expressions of IL-6, TNF-α, tissue factor (TF), VCAM-1, and ICAM-1 were evaluated with qRT-PCR assay and ELISA. Calcein-AM staining was used to determine the attachment of U937 monocytes to HAVECs. quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blot were used to measure the expression level of early growth response 1 (Egr-1) in HAVECs. First, the elevated expression of LOX-1, activated oxidative stress, excessive secreted inflammatory factors, and promoted expression of TF induced by stimulation with ox-LDL were significantly reversed by febuxostat, indicating a protective effect of febuxostat against endothelial dysfunction. Second, the upregulated VCAM-1 and ICAM-1, as well as the increased proportion of adhered monocytes to HAVECs induced by ox-LDL, were significantly alleviated by febuxostat. Finally, the promoted expression level of Egr-1 induced by ox-LDL was pronouncedly suppressed by febuxostat. We conclude that febuxostat protected HAVECs from ox-LDL-induced injury and monocyte attachment.
Collapse
|
18
|
Kushiyama A, Takahashi M, Kushiyama S, Kikuchi T, Asano T. Metabolism-dependent Vascular Pathophysiology in Adult Diseases. YAKUGAKU ZASSHI 2022; 142:465-471. [DOI: 10.1248/yakushi.21-00176-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | - Sakura Kushiyama
- National College of Nursing, National Center for Global Health and Medicine
| | - Takako Kikuchi
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation
| | - Tomoichiro Asano
- Department of Medical Chemistry, Division of Molecular Medical Science, Graduate School of Biomedical Sciences, Hiroshima University
| |
Collapse
|
19
|
Zhu Q, Li XH, Chen HY, Jin QY. The effects of compound centella formula on OxInflammation and silent information regulator 1 in a high-fat diet/streptozotocin-induced diabetic kidney disease rat model. Exp Ther Med 2021; 22:962. [PMID: 34335904 PMCID: PMC8290408 DOI: 10.3892/etm.2021.10394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
The Chinese decoction compound centella formula (CCF) is clinically effective against diabetic kidney disease (DKD), but the exact mechanism remains unclear. The present study aimed to investigate the effects of CCF on OxInflammation and silent information regulator 1 (SIRT1) levels in rats with streptozotocin (STZ)-induced diabetes. Sprague-Dawley rats were divided into CCF, losartan, diabetic control (DC) and normal control (NC) groups (n=7). Except for the NC, all subgroups of rats were fed a high-fat diet for 112 days and received a single intraperitoneal injection of 35 mg/kg STZ on day 29. All rats were sacrificed on day 112. High-performance liquid chromatography was performed to analyse asiaticoside, astragaloside and triptolide levels in CCF (0.3400, 0.0640 and 0.0001 mg/ml, respectively). Fasting blood glucose, urine protein-to-creatinine ratio, serum creatinine and blood urea nitrogen were quantified. Periodic acid Schiff staining, H&E staining and transmission electron microscopy were used to examine kidney pathological changes. The mRNA and protein expression levels of SIRT1 in renal tissues were analysed by reverse transcription-quantitative PCR, western blotting and immunohistochemistry. Oxidative stress was evaluated by measuring the levels of superoxide dismutase (SOD), malondialdehyde (MDA) and nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) in renal tissues. TNF-α and NF-κB p65 subunit in renal tissues were assessed for inflammation. Compared with the rats in the NC group, the rats in the DC group exhibited renal injury with proteinuria, decreased expression levels of SIRT1 and SOD (P<0.01) and increased levels of MDA, NOX4, TNF-α and NF-κB p65 (P<0.01). CCF treatment reduced proteinuria (P<0.01), alleviated renal damage, decreased MDA, NOX4, TNF-α and NF-κB p65 levels (P<0.01), increased SOD levels (P<0.05) and increased SIRT1 mRNA and protein expression levels (P<0.01). The present study indicates that CCF effectively protects the kidney from diabetes by inhibiting OxInflammation and upregulating SIRT1.
Collapse
Affiliation(s)
- Qin Zhu
- Department of Nephrology, Key Laboratory of Zhejiang Province, Management of Kidney Disease, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, P.R. China
| | - Xiao-Hong Li
- Department of Nephrology, Key Laboratory of Zhejiang Province, Management of Kidney Disease, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, P.R. China
| | - Hong-Yu Chen
- Department of Nephrology, Key Laboratory of Zhejiang Province, Management of Kidney Disease, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, P.R. China
| | - Qin-Yang Jin
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
20
|
Nadwa EH, Morcos GNB, Salama NM, Shafik AN. Comparing the Effects of Febuxostat and Allopurinol in an Animal Model of Metabolic Syndrome. Pharmacology 2021; 106:564-572. [PMID: 34182567 DOI: 10.1159/000516495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/01/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Recent studies highlighted the association of hyperuricemia and metabolic syndrome (MS). The aim of this study was to compare the beneficial effects of febuxostat versus allopurinol on the biochemical changes that occur in MS. METHODS Forty adult male Sprague Dawley albino rats were used in the study. Insulin resistance and MS were induced by administration of a high-fructose diet for 8 weeks. Follow-up of changes in weight, blood pressure, serum biochemical parameters, serum antioxidant catalase, and glutathione peroxidase activities was done. At the end of the study, animals were sacrificed, and the thoracic aorta was isolated for in vitro study of the endothelial integrity. RESULTS Allopurinol and febuxostat treatment induced significant reduction in body weight, systolic blood pressure, blood glucose, insulin, lipids, and improved kidney functions and endothelial integrity compared to nontreated rats. Febuxostat was more effective than allopurinol in normalizing serum fasting glucose, uric acid, catalase, and glutathione peroxidase activities. CONCLUSION Xanthine oxidase inhibitors ameliorated the effects of MS. Febuxostat was mildly superior to allopurinol in lowering serum fasting glucose, lipids, uric acid, and antioxidant enzyme activities.
Collapse
Affiliation(s)
- Eman H Nadwa
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Giza, Egypt
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - George N B Morcos
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Cairo University, Giza, Egypt
- Department of Basic Medical Science, Faculty of Medicine, King Salman International University, South Sinai, Egypt
| | - Nagwan M Salama
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Amani N Shafik
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
21
|
Elsisi AEE, Sokar SS, Shalaby MF, Abu-Risha SES. Nephroprotective effects of febuxostat and/or mirtazapine against gentamicin-induced nephrotoxicity through modulation of ERK 1/2, NF-κB and MCP1. Expert Rev Clin Pharmacol 2021; 14:1039-1050. [PMID: 34030558 DOI: 10.1080/17512433.2021.1933435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES This study was conducted to evaluate the potential nephroprotective effects of febuxostat, mirtazapine, and their combination against gentamicin-induced nephrotoxicity. METHODS Induction of nephrotoxicity was achieved via gentamicin injection (100 mg/kg, I.P., for 7 days). Two different doses of mirtazapine (15-30 mg/kg), febuxostat (5-10 mg/kg), and their combination were administered daily for 14 days prior to gentamicin injection and then concomitantly with gentamicin for additional 7 days. Nephrotoxicity was evaluated histopathologically and biochemically. Renal caspase-3, extracellular signal-regulated protein kinase 1/2 (ERK1/2), nuclear factor-kappa-β (NF-κβ), and monocyte chemoattractant protein (MCP-1) were assayed. RESULTS Febuxostat and mirtazapine significantly (p < 0.05) alleviated biochemical and histopathological alterations that were induced by gentamicin and, for the first time, significantly decreased the renal levels of ERK1/2 and MCP-1. Conclusion: Febuxostat and mirtazapine were found to have a synergistic impact in reducing gentamicin-induced nephrotoxicity. EXPERT OPINION The utility of nonpurine xanthine oxidase inhibitor, such as febuxostat and mirtazapine are offering a new potential opportunity for the future nephroprotective effects therapy: Febuxostat and mirtazapine are found to have a synergistic impact in reducing gentamicin-induced nephrotoxicity.
Collapse
Affiliation(s)
| | - Samia Salem Sokar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Marwa Fouad Shalaby
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | | |
Collapse
|
22
|
The Role of Oxidative Stress in Hyperuricemia and Xanthine Oxidoreductase (XOR) Inhibitors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1470380. [PMID: 33854690 PMCID: PMC8019370 DOI: 10.1155/2021/1470380] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022]
Abstract
Uric acid is the end product of purine metabolism in humans. Hyperuricemia is a metabolic disease caused by the increased formation or reduced excretion of serum uric acid (SUA). Alterations in SUA homeostasis have been linked to a number of diseases, and hyperuricemia is the major etiologic factor of gout and has been correlated with metabolic syndrome, cardiovascular disease, diabetes, hypertension, and renal disease. Oxidative stress is usually defined as an imbalance between free radicals and antioxidants in our body and is considered to be one of the main causes of cell damage and the development of disease. Studies have demonstrated that hyperuricemia is closely related to the generation of reactive oxygen species (ROS). In the human body, xanthine oxidoreductase (XOR) catalyzes the oxidative hydroxylation of hypoxanthine to xanthine to uric acid, with the accompanying production of ROS. Therefore, XOR is considered a drug target for the treatment of hyperuricemia and gout. In this review, we discuss the mechanisms of uric acid transport and the development of hyperuricemia, emphasizing the role of oxidative stress in the occurrence and development of hyperuricemia. We also summarize recent advances and new discoveries in XOR inhibitors.
Collapse
|
23
|
Nessa N, Kobara M, Toba H, Adachi T, Yamamoto T, Kanamura N, Pezzotti G, Nakata T. Febuxostat Attenuates the Progression of Periodontitis in Rats. Pharmacology 2021; 106:294-304. [PMID: 33735887 DOI: 10.1159/000513034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/11/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Periodontitis is a lifestyle-related disease that is characterized by chronic inflammation in gingival tissue. Febuxostat, a xanthine oxidase inhibitor, exerts anti-inflammatory and antioxidant effects. OBJECTIVE The present study investigated the effects of febuxostat on periodontitis in a rat model. METHODS Male Wistar rats were divided into 3 groups: control, periodontitis, and febuxostat-treated periodontitis groups. Periodontitis was induced by placing a ligature wire around the 2nd maxillary molar and the administration of febuxostat (5 mg/kg/day) was then initiated. After 4 weeks, alveolar bone loss was assessed by micro-computed tomography and methylene blue staining. The expression of osteoprotegerin (OPG), a bone resorption inhibitor, was detected by quantitative RT-PCR and immunological staining, and the number of osteoclasts in gingival tissue was assessed by tartrate-resistant acid phosphatase staining. The mRNA and protein expression levels of the proinflammatory cytokines, tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β), in gingival tissue were measured using quantitative RT-PCR and immunological staining. Oxidative stress in gingival tissue was evaluated by the expression of 4-hydroxy-2-nonenal (4-HNE), and 8-hydroxy-2-deoxyguanosine (8-OHdG). To clarify the systemic effects of periodontitis, blood pressure and glucose tolerance were examined. RESULTS In rats with periodontitis, alveolar bone resorption was associated with reductions in OPG and increases in osteoclast numbers. The gingival expression of TNF-α, IL-1β, 4-HNE, and 8-OHdG was up-regulated in rats with periodontitis. Febuxostat significantly reduced alveolar bone loss, proinflammatory cytokine levels, and oxidative stress. It also attenuated periodontitis-induced glucose intolerance and blood pressure elevations. CONCLUSION Febuxostat prevented the progression of periodontitis and associated systemic effects by inhibiting proinflammatory mediators and oxidative stress.
Collapse
Affiliation(s)
- Naseratun Nessa
- Division of Pathological Science, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Miyuki Kobara
- Division of Pathological Science, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan,
| | - Hiroe Toba
- Division of Pathological Science, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Giuseppe Pezzotti
- Department of Material Science and Engineering, Kyoto Institute of Technology, Kyoto, Japan
| | - Tetsuo Nakata
- Division of Pathological Science, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
24
|
Hao J, Zhang W, Tong R, Huang Z. Febuxostat Prevents the Cytotoxicity of Propofol in Brain Endothelial Cells. ACS OMEGA 2021; 6:5471-5478. [PMID: 33681587 PMCID: PMC7931401 DOI: 10.1021/acsomega.0c05708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/21/2021] [Indexed: 05/12/2023]
Abstract
Background and purpose: A high risk of brain injury has been reported with the usage of general anesthetics such as propofol in infants. Experimental data indicated that oxidative stress and inflammation are involved in the neurotoxicity induced by propofol. Febuxostat is a novel anti-gout agent recently reported to exert an anti-inflammatory effect. The present study aims to investigate the protective property of febuxostat against the cytotoxicity of propofol in brain endothelial cells as well as the underlying preliminary mechanism. Methods: The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was utilized to screen the optimized incubation concentration of febuxostat. bEnd.3 brain endothelial cells were stimulated with 2% propofol in the presence or absence of febuxostat (10, 20 μM) for 24 h. The lactate dehydrogenase (LDH) release assay was conducted to detect cytotoxicity. The reactive oxygen species (ROS) levels were evaluated using dichloro-dihydro-fluorescein diacetate (DCFH-DA) staining, and the concentration of reduced glutathione (GSH) was determined using a commercial kit. The expressions of TNF-α, IL-6, IL-12, CXCL-1, PDPN, CXCL8, VCAM-1, and E-selectin were determined using a quantitative real-time polymerase chain reaction (qRT-PCR) and an enzyme-linked immunosorbent assay (ELISA). Western blot and qRT-PCR were utilized to determine the expressions of COX-2 and KLF6. The production of PGE2 was evaluated by ELISA. Results: First, increased LDH release induced by propofol was significantly suppressed by febuxostat. The oxidative stress (elevated ROS levels and decreased GSH level) induced by propofol was alleviated by febuxostat. Second, the upregulated inflammatory factors (TNF-α, IL-6, and IL-12), pro-inflammatory chemokines (CXCL-1, PDPN, and CXCL8), adhesion molecules (VCAM-1 and E-selectin), and inflammatory mediators (COX-2 and PGE2) induced by propofol were greatly downregulated by febuxostat. Lastly, the expression of KLF6 was significantly suppressed by propofol but greatly elevated by febuxostat. Conclusion: Febuxostat prevented the cytotoxicity of propofol in brain endothelial cells by alleviating oxidative stress and inflammatory response through KLF6.
Collapse
Affiliation(s)
- Jianli Hao
- Department
of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, PR China
| | - Weiqing Zhang
- Department
of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, PR China
| | - Rui Tong
- Department
of Oncologynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, PR China
| | - Zeqing Huang
- Department
of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, PR China
| |
Collapse
|
25
|
Amelioration of testosterone-induced benign prostatic hyperplasia using febuxostat in rats: The role of VEGF/TGFβ and iNOS/COX-2. Eur J Pharmacol 2020; 889:173631. [PMID: 33031799 DOI: 10.1016/j.ejphar.2020.173631] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 09/23/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
Benign prostatic hyperplasia (BPH) is a common male disorder. Febuxostat is a non-purine, selective inhibitor of xanthine oxidase (XO), which has a strong antioxidant capacity and pleiotropic pharmacological properties. This study's objective was to explore the potential ameliorative effects of febuxostat against testosterone-induced BPH in rats. Febuxostat (10 mg/kg/day, per os [p.o.]) prevented increased prostate index levels, serum levels of prostate-specific antigen (PSA), and testosterone levels compared to animals treated with testosterone alone, when administered for 28 days. Histological examination indicated that febuxostat dramatically ameliorated pathological changes in the prostate architecture compared to the testosterone group. Similarly, febuxostat markedly improved testosterone-induced oxidative stress by inhibiting the increase in lipid peroxide and nitrite content, and by reducing the level of depletion of reduced glutathione (GSH) and superoxide dismutase (SOD) activity, which significantly reduced the prostate content of pro-inflammatory cytokines, including tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6). Furthermore, febuxostat significantly reduced the prostatic content, both in terms of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) messenger ribonucleic acid (mRNA) levels, and of protein levels. Moreover, compared to the testosterone group, febuxostat's beneficial effects prevented the increase in growth factors, comprising vascular endothelial cell growth factor A (VEGF-A) and transforming growth factor beta (TGF-β) protein levels. Its ameliorating effects were equal to those of finasteride, which is the most widely used remedy for BPH. In conclusion, this study provides novel evidence that febuxostat experimentally attenuates testosterone-induced BPH in rats, at least in part by inhibiting iNOS/COX-2 and VEGF/TGF-β pathways.
Collapse
|
26
|
Balakumar P, Alqahtani A, Khan NA, Mahadevan N, Dhanaraj SA. Mechanistic insights into hyperuricemia-associated renal abnormalities with special emphasis on epithelial-to-mesenchymal transition: Pathologic implications and putative pharmacologic targets. Pharmacol Res 2020; 161:105209. [DOI: 10.1016/j.phrs.2020.105209] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023]
|
27
|
Kim YJ, Oh SH, Ahn JS, Yook JM, Kim CD, Park SH, Cho JH, Kim YL. The Crucial Role of Xanthine Oxidase in CKD Progression Associated with Hypercholesterolemia. Int J Mol Sci 2020; 21:ijms21207444. [PMID: 33050202 PMCID: PMC7589966 DOI: 10.3390/ijms21207444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 02/08/2023] Open
Abstract
In the present study, we investigated the effects of xanthine oxidase (XO) inhibition on cholesterol-induced renal dysfunction in chronic kidney disease (CKD) mice, and in low-density lipoprotein (LDL)-treated human kidney proximal tubule epithelial (HK-2) cells. ApoE knockout (KO) mice underwent uninephrectomy to induce CKD, and were fed a normal diet or high-cholesterol (HC) diet along with the XO inhibitor topiroxostat (1 mg/kg/day). HK-2 cells were treated with LDL (200 µg/mL) and topiroxostat (5 µM) or small interfering RNA against xanthine dehydrogenase (siXDH; 20 nM). In uninephrectomized ApoE KO mice, the HC diet increased cholesterol accumulation, oxidative stress, XO activity, and kidney damage, while topiroxostat attenuated the hypercholesterolemia-associated renal dysfunction. The HC diet induced cholesterol accumulation by regulating the expressions of genes involved in cholesterol efflux (Nr1h3 and Abca1) and synthesis (Srebf2 and Hmgcr), which was reversed by topiroxostat. Topiroxostat suppressed the expressions of genes related to hypercholesterolemia-associated inflammation and fibrosis in the unilateral kidney. LDL stimulation evoked changes in the cholesterol metabolism, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and NF-κB pathways in HK-2 cells, which were mitigated by XO inhibition with topiroxostat or siXDH. These findings suggest that XO inhibition exerts renoprotective effects against hypercholesterolemia-associated kidney injury. XO could be a novel therapeutic target for hypercholesterolemia-associated kidney injury in uninephrectomized patients.
Collapse
Affiliation(s)
- You-Jin Kim
- Division of Nephrology, Kyungpook National University Hospital, Daegu 41944, Korea; (Y.-J.K.); (S.-H.O.); (J.-S.A.); (J.-M.Y.); (C.-D.K.); (S.-H.P.)
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea
| | - Se-Hyun Oh
- Division of Nephrology, Kyungpook National University Hospital, Daegu 41944, Korea; (Y.-J.K.); (S.-H.O.); (J.-S.A.); (J.-M.Y.); (C.-D.K.); (S.-H.P.)
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea
| | - Ji-Sun Ahn
- Division of Nephrology, Kyungpook National University Hospital, Daegu 41944, Korea; (Y.-J.K.); (S.-H.O.); (J.-S.A.); (J.-M.Y.); (C.-D.K.); (S.-H.P.)
| | - Ju-Min Yook
- Division of Nephrology, Kyungpook National University Hospital, Daegu 41944, Korea; (Y.-J.K.); (S.-H.O.); (J.-S.A.); (J.-M.Y.); (C.-D.K.); (S.-H.P.)
| | - Chan-Duck Kim
- Division of Nephrology, Kyungpook National University Hospital, Daegu 41944, Korea; (Y.-J.K.); (S.-H.O.); (J.-S.A.); (J.-M.Y.); (C.-D.K.); (S.-H.P.)
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Sun-Hee Park
- Division of Nephrology, Kyungpook National University Hospital, Daegu 41944, Korea; (Y.-J.K.); (S.-H.O.); (J.-S.A.); (J.-M.Y.); (C.-D.K.); (S.-H.P.)
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jang-Hee Cho
- Division of Nephrology, Kyungpook National University Hospital, Daegu 41944, Korea; (Y.-J.K.); (S.-H.O.); (J.-S.A.); (J.-M.Y.); (C.-D.K.); (S.-H.P.)
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (J.-H.C.); (Y.-L.K.); Tel.: +82-10-6566-7551(J.-H.C.); +82-53-420-5553 (Y.-L.K.); Fax: +82-53-426-2046 (J.-H.C.); +82-53-423-7583 (Y.-L.K.)
| | - Yong-Lim Kim
- Division of Nephrology, Kyungpook National University Hospital, Daegu 41944, Korea; (Y.-J.K.); (S.-H.O.); (J.-S.A.); (J.-M.Y.); (C.-D.K.); (S.-H.P.)
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (J.-H.C.); (Y.-L.K.); Tel.: +82-10-6566-7551(J.-H.C.); +82-53-420-5553 (Y.-L.K.); Fax: +82-53-426-2046 (J.-H.C.); +82-53-423-7583 (Y.-L.K.)
| |
Collapse
|
28
|
Maresh MM, Abdelaziz RR, Ibrahim TM. Febuxostat mitigates concanavalin A-induced acute liver injury via modulation of MCP-1, IL-1β, TNF-α, neutrophil infiltration, and apoptosis in mice. Life Sci 2020; 260:118307. [PMID: 32841665 DOI: 10.1016/j.lfs.2020.118307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/10/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022]
Abstract
AIM Liver plays a crucial role in innate immunity reactions. This role predisposes the liver to innate-mediated liver injury when uncontrolled inflammation occurs. In this study, the effect of febuxostat administration on acute liver injury induced by concanavalin A (Con A) injection into mouse eye orbital sinus was studied. MATERIALS AND METHODS Two doses of febuxostat (10 and 20 mg/kg, orally) were administered either 1 h before or 30 min after the administration of Con A. Febuxostat at a low dose (10 mg/kg) before and after Con A modulated the elevation of serum ALT, liver uric acid, liver myeloperoxidase (MPO), and interleukin-1β (IL-1β) induced by Con A. The same dose of febuxostat before Con A also decreased serum total bilirubin and neutrophil infiltration, as evidenced by flow cytometry and histopathological analysis. KEY FINDINGS Febuxostat at a high dose (20 mg/kg) significantly improved serum ALT, AST, albumin, total bilirubin, liver uric acid, MPO, monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), interleukin-4 (IL-4), IL-1β, and neutrophil infiltration induced by Con A administration. The results of histopathological examination of liver cells paralleled the observed biochemical improvements. Hepatocyte apoptosis as evidenced by immunohistochemical examination of cleaved caspase-3 was markedly decreased in the febuxostat protection and treatment groups, in a dose-dependent manner SIGNIFICANCE: These results indicate that febuxostat, especially at the higher dose, may be an effective inhibitor of immune reactions evoked by Con A administration.
Collapse
Affiliation(s)
- Mohammed M Maresh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt..
| | - Tarek M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| |
Collapse
|
29
|
Deng H, Zhang BL, Tong JD, Yang XH, Jin HM. Febuxostat Use and Risks of Cardiovascular Disease Events, Cardiac Death, and All-cause Mortality: Metaanalysis of Randomized Controlled Trials. J Rheumatol 2020; 48:1082-1089. [PMID: 32801136 DOI: 10.3899/jrheum.200307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To assess whether febuxostat use increases the risk of developing cardiovascular (CV) events, cardiac death, and all-cause mortalities. METHODS The relevant literature was searched in several databases including MEDLINE (PubMed, January 1, 1966-February 29, 2020), Web of Science, EMBASE (January 1, 1974-February 29, 2020), ClinicalTrials. gov, and Cochrane Central Register of Controlled Trials. Manual searches for references cited in the original studies and relevant review articles were also performed. All studies included in this metaanalysis were published in English. RESULTS In the end, 20 studies that met our inclusion criteria were included in our metaanalysis. Use of febuxostat was found not to be associated with an increased risk of all-cause mortality (RR 0.87, 95% CI 0.57-1.32, P = 0.51). Also, there was no association between febuxostat use and mortalities arising from CV diseases (CVD; RR 0.84, 95% CI 0.49-1.45, P = 0.53). The RR also revealed that febuxostat use was not associated with CVD events (RR 0.98, 95% CI 0.83-1.16, P = 0.83). Further, the likelihood of occurrence of CVD events was found not to be dependent on febuxostat dose (RR 1.04, 95% CI 0.84-1.30, P = 0.72). CONCLUSION Febuxostat use is not associated with increased risks of all-cause mortality, death from CVD, or CVD events. Accordingly, it is a safe drug for the treatment of gout.
Collapse
Affiliation(s)
- Hao Deng
- H. Deng, MD, X.H. Yang, MD, H.M. Jin, PhD, Division of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center
| | - Bao Long Zhang
- B.L. Zhang, PhD, The Institutes of Biomedical Sciences (IBS), Fudan University
| | - Jin Dong Tong
- J.D. Tong, PhD, Division of Vascular Surgery, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China.
| | - Xiu Hong Yang
- H. Deng, MD, X.H. Yang, MD, H.M. Jin, PhD, Division of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center;
| | - Hui Min Jin
- H. Deng, MD, X.H. Yang, MD, H.M. Jin, PhD, Division of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center;
| |
Collapse
|
30
|
Omizo H, Tamura Y, Morimoto C, Ueno M, Hayama Y, Kuribayashi-Okuma E, Uchida S, Shibata S. Cardio-renal protective effect of the xanthine oxidase inhibitor febuxostat in the 5/6 nephrectomy model with hyperuricemia. Sci Rep 2020; 10:9326. [PMID: 32518351 PMCID: PMC7283314 DOI: 10.1038/s41598-020-65706-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023] Open
Abstract
Although hyperuricemia has been shown to be associated with the progression of cardiovascular disorder and chronic kidney disease (CKD), there is conflicting evidence as to whether xanthine oxidase (XO) inhibitors confer organ protection besides lowering serum urate levels. In this study, we addressed the cardio-renal effects of XO inhibition in rodent CKD model with hyperuricemia. Sprague-Dawley rats underwent 5/6 nephrectomy and received a uricase inhibitor oxonic acid for 8 weeks (RK + HUA rats). In some rats, a XO inhibitor febuxostat was administered orally. Compared with control group, RK + HUA group showed a significant increase in albuminuria and renal injury. Febuxostat reduced serum uric acid as well as urinary albumin levels. Histological and immunohistochemical analysis of the kidney revealed that febuxostat alleviated glomerular, tubulointerstitial, and arteriolar injury in RK + HUA rats. Moreover, in the heart, RK + HUA showed individual myofiber hypertrophy and cardiac fibrosis, which was significantly attenuated by febuxostat. We found that renal injury and the indices of cardiac changes were well correlated, confirming the cardio-renal interaction in this model. Finally, NF-E2-related factor 2 (Nrf2) and the downstream target heme oxygenase-1 (HO-1) protein levels were increased both in the heart and in the kidney in RK + HUA rats, and these changes were alleviated by febuxostat, suggesting that tissue oxidative stress burden was attenuated by the treatment. These data demonstrate that febuxostat protects against cardiac and renal injury in RK + HUA rats, and underscore the pathological importance of XO in the cardio-renal interaction.
Collapse
Affiliation(s)
- Hiroki Omizo
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Yoshifuru Tamura
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.
| | - Chikayuki Morimoto
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Masaki Ueno
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Yuto Hayama
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Emiko Kuribayashi-Okuma
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Shunya Uchida
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.,Department of Health Care, Teikyo Heisei University, 2-51-4 Higashi-Ikebukuro, Toshima-ku, Tokyo, 170-8445, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.
| |
Collapse
|
31
|
Regulation of Vascular Function and Inflammation via Cross Talk of Reactive Oxygen and Nitrogen Species from Mitochondria or NADPH Oxidase-Implications for Diabetes Progression. Int J Mol Sci 2020; 21:ijms21103405. [PMID: 32408480 PMCID: PMC7279344 DOI: 10.3390/ijms21103405] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress plays a key role for the development of cardiovascular, metabolic, and neurodegenerative disease. This concept has been proven by using the approach of genetic deletion of reactive oxygen and nitrogen species (RONS) producing, pro-oxidant enzymes as well as by the overexpression of RONS detoxifying, antioxidant enzymes leading to an amelioration of the severity of diseases. Vice versa, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of RONS detoxifying enzymes. We have previously identified cross talk mechanisms between different sources of RONS, which can amplify the oxidative stress-mediated damage. Here, the pathways and potential mechanisms leading to this cross talk are analyzed in detail and highlighted by selected examples from the current literature and own data including hypoxia, angiotensin II (AT-II)-induced hypertension, nitrate tolerance, aging, and others. The general concept of redox-based activation of RONS sources via “kindling radicals” and enzyme-specific “redox switches” as well as the interaction with redox-sensitive inflammatory pathways are discussed. Here, we present evidence for the existence of such cross talk mechanisms in the setting of diabetes and critically assess their contribution to the severity of diabetic complications.
Collapse
|
32
|
Baer PC, Koch B, Geiger H. Kidney Inflammation, Injury and Regeneration. Int J Mol Sci 2020; 21:ijms21031164. [PMID: 32050569 PMCID: PMC7036853 DOI: 10.3390/ijms21031164] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
Damage to kidney cells can occur due to a variety of ischemic and toxic insults and leads to inflammation and cell death, which can result in acute kidney injury (AKI) [...].
Collapse
Affiliation(s)
- Patrick C. Baer
- Correspondence: ; Tel.: +49-69-6301-5554; Fax: +49-69-6301-4749
| | | | | |
Collapse
|