1
|
Dong M, Cui Z, Liu Y, Bu Y, An K, Mao L. Effects of Febuxostat Therapy on Circulating Adipokine Profiles in Patients with Overweight or Obesity and Asymptomatic Hyperuricemia: A Randomized Controlled Study. Obes Facts 2024; 17:524-534. [PMID: 39116844 PMCID: PMC11458164 DOI: 10.1159/000540701] [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: 04/19/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
INTRODUCTION Elevated levels of serum uric acid (SUA) are strongly associated with several components of the metabolic syndrome, particularly obesity. Previous studies have reported the correlation between SUA levels, xanthine oxidoreductase (XOR) activity, and the imbalanced adipokine levels that are characteristic of obesity. In this study, we explored the effect of febuxostat on circulating adipokine profiles in patients with overweight or obesity and asymptomatic hyperuricemia. METHODS This study was a single-center, randomized, and controlled clinical trial that enrolled 130 participants with asymptomatic hyperuricemia and obesity. One hundred seventeen participants were included in the final analysis, with 60 participants in the febuxostat group and 57 in the control group. We compared the circulating adipokine levels at 3 and 6 months, including high molecular weight (HMW) adiponectin, chemerin, omentin, monocyte chemotactic protein-1, asprosin, fibroblast growth factor 21, neuregulin-4, leptin, resistin, vaspin, visfatin, adipsin, and assessed the correlation between changes in adipokine levels (Δadipokines) and changes in XOR activity (ΔXOR) after febuxostat treatment. RESULTS The results showed that an increase in HMW adiponectin and omentin levels and a decrease in chemerin and asprosin levels at 3 or 6 months compared to the control group. Additionally, a positive correlation was observed between ΔXOR activity and Δasprosin. Furthermore, after adjusting for triglyceride (ΔTG) and serum uric acid (ΔSUA) in multiple linear regression analyses, we found that ΔXOR activity was independently correlated with Δasprosin. CONCLUSION This study may provide important evidence that febuxostat could alleviate the imbalance in circulating adipokine levels in patients with overweight or obesity and asymptomatic hyperuricemia. Furthermore, we observed a positive correlation between changes in asprosin levels and changes in XOR activity after febuxostat treatment.
Collapse
Affiliation(s)
- Meijuan Dong
- Department of Endocrinology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Zhaohui Cui
- Department of Endocrinology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Yuanyuan Liu
- Department of Endocrinology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Yanlong Bu
- Department of Endocrinology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Kun An
- Department of Neurology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Li Mao
- Department of Endocrinology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
2
|
Zeng L, Shali S, Gao Y, Du X, Zhu X, Li L, Dai Y, Zhou P. CRISPR/Cas9 Mediated Deletion of the Uox Gene Generates a Mouse Model of Hyperuricemia with Multiple Complications. J Cardiovasc Transl Res 2024:10.1007/s12265-024-10526-6. [PMID: 38856882 DOI: 10.1007/s12265-024-10526-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024]
Abstract
Hyperuricemia is a common metabolic disorder with severe complications. We aimed to develop a mouse model for spontaneous hyperuricemia. Uox-/- mouse model was generated on C57BL/6J background by deleting exon 2-4 of Uox using the CRISPR/Cas9 system. The prototypic Uox -/-mice had 5.5-fold increased serum uric acid (1351.04±276.58μmol/L) as compared to the wild type mice (P<0.0001), but died by 4 weeks. After allopurinol (3ug/g) intervention, they all survived > 8 weeks. The serum uric acid was 612.55±146.98μmol/L in the 8-week-old allopurinol-rescued Uox -/-mice, which manifested multiple complications including severe renal insufficiency, hypertension, left ventricular remodeling and systolic dysfunction, aortic endothelial dysfunction, hepatic steatosis and elevated liver enzymes, as well as hyperglycemia and hypercholesteremia. The present Uox-/- mice developed spontaneous hyperuricemia complicated with urate nephropathy, cardiovascular disease and cardiometabolic disorders, and may provide a novel tool to study hyperuricemia associated early-onset cardiovascular disorders in human.
Collapse
Affiliation(s)
- Linzi Zeng
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shalaimaiti Shali
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, China.
| | - Yabiao Gao
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xingchen Du
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Xiaoxia Zhu
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Lin Li
- Department of Nephrology, Shanghai Changzheng Hospital, The Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yuxiang Dai
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, China.
| | - Ping Zhou
- Department of Physiology and Pathophysiology of School of Basic Medical Sciences, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Cao P, Yue M, Cheng Y, Sullivan MA, Chen W, Yu H, Li F, Wu S, Lv Y, Zhai X, Zhang Y. Naringenin prevents non-alcoholic steatohepatitis by modulating the host metabolome and intestinal microbiome in MCD diet-fed mice. Food Sci Nutr 2023; 11:7826-7840. [PMID: 38107095 PMCID: PMC10724642 DOI: 10.1002/fsn3.3700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 12/19/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a severe inflammatory phase of the non-alcoholic fatty liver disease (NAFLD) spectrum and can progress to advanced stages of NAFLD if left untreated. This study uses multi-omics data to elucidate the underlying mechanism of naringenin's reported benefit in alleviating (NASH). Male mice were fed a NASH-inducing (methionine-choline-deficient) MCD diet with or without naringenin supplementation for 6 weeks. Naringenin prevented NASH-induced histopathological liver damage and reversed the abnormal levels of hepatic triglyceride (TG)/total cholesterol (TC), serum TG/TC, serum alanine aminotransferase/aspartate transaminase, and hepatic malondialdehyde and glutathione. Importantly, naringenin intervention significantly modulated the relative abundance of gut microbiota and the host metabolomic profile. We detected more than 700 metabolites in the serum and found that the gut genus levels of Anaeroplasma and the [Eubacterium] nodatum group were closely associated with xanthine, 2-picoline, and securinine, respectively. Tuzzerella alterations showed the highest number of associations with host endogenous metabolites such as FAHFA (8:0/10:0), FFA (20:2), carnitine C8:1, tridecanedioic acid, securinine, acetylvaline, DL-O-tyrosine, and Phe-Asn. This study indicates that the interplay between host serum metabolites and gut microbiota may contribute to the therapeutic effect of naringenin against NASH.
Collapse
Affiliation(s)
- Peng Cao
- Department of Pharmacy, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Clinical Research Center for Precision Medicine for Critical IllnessWuhanChina
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical SciencesHubei University of MedicineShiyanChina
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ming Yue
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical SciencesHubei University of MedicineShiyanChina
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuanlei Cheng
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical SciencesHubei University of MedicineShiyanChina
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Mitchell A. Sullivan
- Glycation and Diabetes, Mater Research Institute – The University of QueenslandTranslational Research InstituteBrisbaneQueenslandAustralia
| | - Wen Chen
- Department of Pharmacy, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Clinical Research Center for Precision Medicine for Critical IllnessWuhanChina
| | - Huifan Yu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical SciencesHubei University of MedicineShiyanChina
| | - Fei Li
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical SciencesHubei University of MedicineShiyanChina
| | - Sanlan Wu
- Department of Pharmacy, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Clinical Research Center for Precision Medicine for Critical IllnessWuhanChina
| | - Yongning Lv
- Department of Pharmacy, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Clinical Research Center for Precision Medicine for Critical IllnessWuhanChina
| | - Xuejia Zhai
- Department of Pharmacy, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Clinical Research Center for Precision Medicine for Critical IllnessWuhanChina
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Clinical Research Center for Precision Medicine for Critical IllnessWuhanChina
| |
Collapse
|
4
|
Al-Shargi A, El Kholy AA, Adel A, Hassany M, Shaheen SM. Allopurinol versus Febuxostat: A New Approach for the Management of Hepatic Steatosis in Metabolic Dysfunction-Associated Steatotic Liver Disease. Biomedicines 2023; 11:3074. [PMID: 38002074 PMCID: PMC10669273 DOI: 10.3390/biomedicines11113074] [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: 09/20/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) includes patients with hepatic steatosis and at least one of five cardiometabolic risk factors. Xanthine oxidase (XO) represents a treatment target for MASLD. We aimed to evaluate the effect of two xanthine oxidase inhibitors, allopurinol and febuxostat, plus lifestyle modifications compared to lifestyle modifications alone on improving steatosis. Ninety MASLD patients were assigned to one of three groups for three months. Patients with hyperuricemia were given either allopurinol 100 mg or febuxostat 40 mg daily, along with lifestyle modifications. The third control group was only given lifestyle modifications, excluding all patients with hyperuricemia due to ethical concerns. The primary outcome was to measure the change in the controlled attenuation parameter (CAP) score as an indicator of steatosis from baseline after three months. The secondary outcome was to measure the change in serum uric acid (SUA) three months from baseline. The study found that the CAP score decreased significantly in the allopurinol group (p = 0.009), but the decline in the febuxostat or lifestyle groups was non-significant (p = 0.189 and 0.054, respectively). The SUA levels were significantly reduced in both the allopurinol and febuxostat groups (p < 0.001), with no statistical difference between the two groups (p = 0.496).
Collapse
Affiliation(s)
- Amani Al-Shargi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 4393005, Egypt; (A.A.E.K.); (S.M.S.)
| | - Amal A. El Kholy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 4393005, Egypt; (A.A.E.K.); (S.M.S.)
| | - Abdulmoneim Adel
- National Hepatology and Tropical Medicine Research Institute (NHTMRI), Cairo 4260010, Egypt; (A.A.); (M.H.)
| | - Mohamed Hassany
- National Hepatology and Tropical Medicine Research Institute (NHTMRI), Cairo 4260010, Egypt; (A.A.); (M.H.)
| | - Sara M. Shaheen
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 4393005, Egypt; (A.A.E.K.); (S.M.S.)
| |
Collapse
|
5
|
Dang SW, Gao L, Li YJ, Zhang R, Xu J. Metabolic characteristics of non-obese and obese metabolic dysfunction-associated fatty liver disease in type 2 diabetes mellitus and its association with diabetic peripheral neuropathy and diabetic retinopathy. Front Med (Lausanne) 2023; 10:1216412. [PMID: 37828942 PMCID: PMC10566373 DOI: 10.3389/fmed.2023.1216412] [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: 06/08/2023] [Accepted: 08/28/2023] [Indexed: 10/14/2023] Open
Abstract
Aim To assess the metabolic characteristics of non-obese metabolic dysfunction-associated fatty liver disease (MAFLD) compared with obese MAFLD and the relationship of MAFLD with diabetic peripheral neuropathy and diabetic retinopathy in patients with Type 2 diabetes mellitus (T2DM). Methods Data were obtained from 536 T2DM patients (355 women, 181 men; age 58.2 ± 12.0 years). We explored the difference in clinical characteristics between obese MAFLD (body mass index ≥25 kg/m2) and non-obese MAFLD (body mass index <25 kg/m2) in T2DM patients. One-way analysis of variance (ANOVA) was used to compare the means of continuous variables, and the Chi-squared test was used to compare the differences in frequencies of categorical variables. Logistic regression models were adopted to calculate odds ratios. Results The prevalence of MAFLD in hospitalized Chinese T2DM patients was calculated to be 42.7%. Both obese and non-obese MAFLD patients had higher levels of body mass index (BMI), waist circumfere nce, triglyceride, alanine aminotransferase, aspar tate aminotransferase, γ-glutamyltransferase, you nger age, higher prevalence of hyperlipidemia and shorter duration of T2DM and lower incidence of diabetic retinopathy, compared with participants with out MAFLD in the same weight group. Uric acid levels were positively correlated with the risk of MAFLD only in non-obese subjects but not in obese subjects. In non-obese patients with T2DM, a negative correlation was found between the prevalence of MAFLD and diabetic retinopathy. Conclusion Even in non-obese patients with T2DM, BMI was found to be an independent risk factor for MAFLD. These findings support a more structured, risk-factor-based approach to MAFLD management, particularly in patients with T2DM. Non-obese MAFLD has unique results in metabolic characteristics and the correlation with diabetic retinopathy and diabetic peripheral neuropathy, which should be further explored.
Collapse
Affiliation(s)
- Si-Wen Dang
- Department of Endocrinology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- International Center for Obesity and Metabolic Disease Research, Xi’an Jiaotong University, Xi’an, China
| | - Lei Gao
- Department of Endocrinology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- International Center for Obesity and Metabolic Disease Research, Xi’an Jiaotong University, Xi’an, China
| | - Yu-Jun Li
- Department of Endocrinology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- International Center for Obesity and Metabolic Disease Research, Xi’an Jiaotong University, Xi’an, China
| | - Ruo Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- International Center for Obesity and Metabolic Disease Research, Xi’an Jiaotong University, Xi’an, China
| | - Jing Xu
- Department of Endocrinology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- International Center for Obesity and Metabolic Disease Research, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
6
|
Feng Y, Zheng S, Liu L, Yang Y. Association of serum uric acid with hepatic steatosis detected by controlled attenuation parameter in the United States population. Lipids Health Dis 2023; 22:76. [PMID: 37340407 DOI: 10.1186/s12944-023-01846-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/09/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND The relationship between serum uric acid (SUA) and nonalcoholic fatty liver disease (NAFLD) has been previously reported. Controlled attenuation parameter (CAP) has better diagnostic performance than ultrasonography for assessing hepatic steatosis. The association of SUA with hepatic steatosis detected by CAP is worth further study. METHODS The US population aged 20 years or older from the National Health and Nutrition Examination Survey (NHANES) was assessed. Hepatic steatosis was evaluated by the controlled attenuation parameter (CAP). NAFLD status was defined as CAP values of 268 dB/m without hepatitis B or C virus infection or considerable alcohol consumption. Multiple imputations were performed to fill in the missing covariate values. Linear regression, logistic regression, and smooth curve fitting were used to examine the association. RESULTS In total, 3919 individuals participated in this study. There was a positive association between SUA (µmol/L) and CAP (β = 0.14, 95% CI: 0.12-0.17, P < 0.01). After stratification by sex, a significant relationship between SUA and CAP existed in both males (β = 0.12, 95% CI: 0.09-0.16, P < 0.01) and females (β = 0.17, 95% CI: 0.14-0.20, P < 0.01) after multiple imputation. The inflection points of the threshold effect of SUA on CAP were 487.7 µmol/L in males and 386.6 µmol/L in females. There was a positive association between SUA (mg/dL) and NAFLD (OR = 1.30, 95% CI: 1.23-1.37, P < 0.01). After stratification by race, positive relationships were also observed. Meanwhile, a positive relationship existed between hyperuricemia and NAFLD (OR = 1.94, 95% CI: 1.64-2.30, P < 0.01). The positive relationship was more significant in females than in males (P for interaction < 0.01). CONCLUSIONS There was a positive association between SUA and CAP, as well as between SUA and NAFLD. Subgroup studies stratified by sex and ethnicity demonstrated that the effects were consistent.
Collapse
Affiliation(s)
- Yunfu Feng
- Endoscopy Center, The First People's Hospital of Kunshan, Kunshan, 215300, China
| | - Sijie Zheng
- Endoscopy Center, The First People's Hospital of Kunshan, Kunshan, 215300, China
| | - Luojie Liu
- Department of Gastroenterology, Changshu Hospital Affiliated to Soochow University, Changshu, 215500, China.
| | - Yanting Yang
- Department of Gastroenterology, The Third People's Hospital of Kunshan, Kunshan, 215300, China.
| |
Collapse
|
7
|
Kakimoto M, Fujii M, Sato I, Honma K, Nakayama H, Kirihara S, Fukuoka T, Ran S, Hirohata S, Kitamori K, Yamamoto S, Watanabe S. Antioxidant action of xanthine oxidase inhibitor febuxostat protects the liver and blood vasculature in SHRSP5/Dmcr rats. J Appl Biomed 2023; 21:80-90. [PMID: 37376883 DOI: 10.32725/jab.2023.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/25/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Xanthine oxidase (XO) generates reactive oxygen species during uric acid production. Therefore, XO inhibitors, which suppress oxidative stress, may effectively treat non-alcoholic steatohepatitis (NASH) and atherosclerosis via uric acid reduction. In this study, we examined the antioxidant effect of the XO inhibitor febuxostat on NASH and atherosclerosis in stroke-prone spontaneously hypertensive 5 (SHRSP5/Dmcr) rats. METHODS SHRSP5/Dmcr rats were divided into three groups: SHRSP5/Dmcr + high-fat and high-cholesterol (HFC) diet [control group, n = 5], SHRSP5/Dmcr + HFC diet + 10% fructose (40 ml/day) [fructose group, n = 5], and SHRSP5/Dmcr + HFC diet + 10% fructose (40 ml/day) + febuxostat (1.0 mg/kg/day) [febuxostat group, n = 5]. Glucose and insulin resistance, blood biochemistry, histopathological staining, endothelial function, and oxidative stress markers were evaluated. RESULTS Febuxostat reduced the plasma uric acid levels. Oxidative stress-related genes were downregulated, whereas antioxidant factor-related genes were upregulated in the febuxostat group compared with those in the fructose group. Febuxostat also ameliorated inflammation, fibrosis, and lipid accumulation in the liver. Mesenteric lipid deposition decreased in the arteries, and aortic endothelial function improved in the febuxostat group. CONCLUSIONS Overall, the XO inhibitor febuxostat exerted protective effects against NASH and atherosclerosis in SHRSP5/Dmcr rats.
Collapse
Affiliation(s)
- Mai Kakimoto
- Okayama University, Graduate School of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Moe Fujii
- Okayama University, Graduate School of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Ikumi Sato
- Okayama University, Graduate School of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Koki Honma
- Okayama University, Graduate School of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Hinako Nakayama
- Okayama University, Graduate School of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Sora Kirihara
- Okayama University, Graduate School of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Taketo Fukuoka
- Okayama University, Faculty of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Shang Ran
- Okayama University, Graduate School of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Satoshi Hirohata
- Okayama University, Academic Field of Health Science, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Kazuya Kitamori
- Kinjo Gakuin University, College of Human Life and Environment, 2-1723, Omori, Moriyama-ku, Nagoya-shi, Aichi, 463-8521, Japan
| | - Shusei Yamamoto
- Okayama University, Graduate School of Health Sciences, Department of Medical Technology, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
- Okayama University, Academic Field of Health Science, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Shogo Watanabe
- Okayama University, Academic Field of Health Science, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| |
Collapse
|
8
|
Khrais A, Kahlam A, Tahir A, Shaikh A, Ahlawat S. Outcomes of gout in patients with cirrhosis: A national inpatient sample-based study. World J Hepatol 2023; 15:303-310. [PMID: 36926244 PMCID: PMC10011910 DOI: 10.4254/wjh.v15.i2.303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/06/2023] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Hyperuricemia is a prerequisite for the development of gout. Elevated serum uric acid (UA) levels result from either overproduction or decreased excretion. A positive correlation between serum UA levels, cirrhosis-related complications and the incidence of nonalcoholic fatty liver disease has been established, but it is unknown whether hyperuricemia results in worsening cirrhosis outcomes. We hypothesize that patients with cirrhosis will have poorer gout outcomes.
AIM To explore the link between cirrhosis and the incidence of gout-related complications.
METHODS This was a cross-sectional study. The national inpatient sample was used to identify patients hospitalized with gout, stratified based on a history of cirrhosis, from 2001 to 2013 via the International Classification of Diseases, Ninth Revision, Clinical Modification codes. Primary outcomes were mortality, gout complications and joint interventions. The χ2 test and independent t-test were performed to assess categorical and continuous data, respectively. Multiple logistic regression was used to control for confounding variables.
RESULTS Patients without cirrhosis were older (70.37 ± 13.53 years vs 66.21 ± 12.325 years; P < 0.05). Most patients were male (74.63% in the cirrhosis group vs 66.83%; adjusted P < 0.05). Patients with cirrhosis had greater rates of mortality (5.49% vs 2.03%; adjusted P < 0.05), gout flare (2.89% vs 2.77%; adjusted P < 0.05) and tophi (0.97% vs 0.75%; adjusted P = 0.677). Patients without cirrhosis had higher rates of arthrocentesis (2.45% vs 2.21%; adjusted P < 0.05) and joint injections (0.72% vs 0.52%; adjusted P < 0.05).
CONCLUSION Gout complications were more common in cirrhosis. Those without cirrhosis had higher rates of interventions, possibly due to hesitancy with performing these interventions given the higher complication risk in cirrhosis.
Collapse
Affiliation(s)
- Ayham Khrais
- Division of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Aaron Kahlam
- Division of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Ali Tahir
- Division of Medicine, St. Luke’s University Health Network, Bethlehem, PA 18015, United States
| | - Amjad Shaikh
- Division of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Sushil Ahlawat
- Division of Gastroenterology and Hepatology, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| |
Collapse
|
9
|
The Effects of Topiroxostat, a Selective Xanthine Oxidoreductase Inhibitor, on Arterial Stiffness in Hyperuricemic Patients with Liver Dysfunction: A Sub-Analysis of the BEYOND-UA Study. Biomedicines 2023; 11:biomedicines11030674. [PMID: 36979653 PMCID: PMC10045538 DOI: 10.3390/biomedicines11030674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Background: The effects of uric acid (UA)-lowering therapy with xanthine oxidoreductase (XOR) inhibitors on the development of cardiovascular diseases remain controversial. Based on recent findings that plasma XOR activity increased in liver disease conditions, we conducted a sub-analysis of the BEYOND-UA study to examine the differential effects of topiroxostat on arterial stiffness based on liver function in hyperuricemic individuals with hypertension. Methods: Sixty-three subjects treated with topiroxostat were grouped according to baseline alanine aminotransferase (ALT) levels (above or below cut-off values of 22, 30, or 40 U/L). The primary endpoint was changes in the cardio-ankle vascular index (CAVI) from baseline to 24 weeks. Results: Significant reductions in CAVI during topiroxostat therapy occurred in subjects with baseline ALT ≥30 U/L or ≥40 U/L, and significant between-group differences were detected. Brachial-ankle pulse wave velocity significantly decreased in the ALT-high groups at all cut-off values. Reductions in morning home blood pressure and serum UA were similar regardless of the baseline ALT level. For eleven subjects with available data, ALT-high groups showed high plasma XOR activity, which was significantly suppressed by topiroxostat. Conclusions: Topiroxostat improved arterial stiffness parameters in hyperuricemic patients with liver dysfunction, which might be related to its inhibitory effect on plasma XOR.
Collapse
|
10
|
Keles U, Ow JR, Kuentzel KB, Zhao LN, Kaldis P. Liver-derived metabolites as signaling molecules in fatty liver disease. Cell Mol Life Sci 2022; 80:4. [PMID: 36477411 PMCID: PMC9729146 DOI: 10.1007/s00018-022-04658-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
Excessive fat accumulation in the liver has become a major health threat worldwide. Unresolved fat deposition in the liver can go undetected until it develops into fatty liver disease, followed by steatohepatitis, fibrosis, cirrhosis, and eventually hepatocellular carcinoma. Lipid deposition in the liver is governed by complex communication, primarily between metabolic organs. This can be mediated by hormones, organokines, and also, as has been more recently discovered, metabolites. Although how metabolites from peripheral organs affect the liver is well documented, the effect of metabolic players released from the liver during the development of fatty liver disease or associated comorbidities needs further attention. Here we focus on interorgan crosstalk based on metabolites released from the liver and how these molecules act as signaling molecules in peripheral tissues. Due to the liver's specific role, we are covering lipid and bile mechanism-derived metabolites. We also discuss the high sucrose intake associated with uric acid release from the liver. Excessive fat deposition in the liver during fatty liver disease development reflects disrupted metabolic processes. As a response, the liver secretes a variety of signaling molecules as well as metabolites which act as a footprint of the metabolic disruption. In the coming years, the reciprocal exchange of metabolites between the liver and other metabolic organs will gain further importance and will help to better understand the development of fatty liver disease and associated diseases.
Collapse
Affiliation(s)
- Umur Keles
- Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden
| | - Jin Rong Ow
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Katharina Barbara Kuentzel
- Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden
| | - Li Na Zhao
- Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden
| | - Philipp Kaldis
- Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden. .,Lund University Diabetes Centre (LUDC), Clinical Research Centre (CRC), Lund University, Box 50332, 202 13, Malmö, Sweden.
| |
Collapse
|
11
|
Lewis SE, Li L, Fazzari M, Salvatore SR, Li J, Hileman EA, Maxwell BA, Schopfer FJ, Arteel GE, Khoo NK, Kelley EE. Obese female mice do not exhibit overt hyperuricemia despite hepatic steatosis and impaired glucose tolerance. ADVANCES IN REDOX RESEARCH : AN OFFICIAL JOURNAL OF THE SOCIETY FOR REDOX BIOLOGY AND MEDICINE AND THE SOCIETY FOR FREE RADICAL RESEARCH-EUROPE 2022; 6:100051. [PMID: 36561324 PMCID: PMC9770588 DOI: 10.1016/j.arres.2022.100051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent reports have clearly demonstrated a tight correlation between obesity and elevated circulating uric acid levels (hyperuricemia). However, nearly all preclinical work in this area has been completed with male mice, leaving the field with a considerable gap in knowledge regarding female responses to obesity and hyperuricemia. This deficiency in sex as a biological variable extends beyond unknowns regarding uric acid (UA) to several important comorbidities associated with obesity including nonalcoholic fatty liver disease (NAFLD). To attempt to address this issue, herein we describe both phenotypic and metabolic responses to diet-induced obesity (DIO) in female mice. Six-week-old female C57BL/6J mice were fed a high-fat diet (60% calories derived from fat) for 32 weeks. The DIO female mice had significant weight gain over the course of the study, higher fasting blood glucose, impaired glucose tolerance, and elevated plasma insulin levels compared to age-matched on normal chow. While these classic indices of DIO and NAFLD were observed such as increased circulating levels of ALT and AST, there was no difference in circulating UA levels. Obese female mice also demonstrated increased hepatic triglyceride (TG), cholesterol, and cholesteryl ester. In addition, several markers of hepatic inflammation were significantly increased. Also, alterations in the expression of redox-related enzymes were observed in obese mice compared to lean controls including increases in extracellular superoxide dismutase (Sod3), heme oxygenase (Ho)-1, and xanthine dehydrogenase (Xdh). Interestingly, hepatic UA levels were significantly elevated (~2-fold) in obese mice compared to their lean counterparts. These data demonstrate female mice assume a similar metabolic profile to that reported in several male models of obesity in the context of alterations in glucose tolerance, hepatic steatosis, and elevated transaminases (ALT and AST) in the absence of hyperuricemia affirming the need for further study.
Collapse
Affiliation(s)
- Sara E. Lewis
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, 3072B Health Sciences Center, PO Box 9229, Morgantown, WV 26506-9229, USA
| | - Lihua Li
- Department of Pharmacology & Chemical Biology, USA
| | | | | | - Jiang Li
- Division of Gastroenterology, Hepatology and Nutrition, USA
| | - Emily A. Hileman
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, 3072B Health Sciences Center, PO Box 9229, Morgantown, WV 26506-9229, USA
| | - Brooke A. Maxwell
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, 3072B Health Sciences Center, PO Box 9229, Morgantown, WV 26506-9229, USA
| | - Francisco J. Schopfer
- Department of Pharmacology & Chemical Biology, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Gavin E. Arteel
- Division of Gastroenterology, Hepatology and Nutrition, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nicholas K.H. Khoo
- Department of Pharmacology & Chemical Biology, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Corresponding author at: Department of Pharmacology & Chemical Biology, University of Pittsburgh, 200 Lothrop Street, E1340 Thomas E. Starzl Biomedical Science Tower, Pittsburgh, PA 15261, (N.K.H. Khoo)
| | - Eric E. Kelley
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, 3072B Health Sciences Center, PO Box 9229, Morgantown, WV 26506-9229, USA
- Corresponding author: (E.E. Kelley)
| |
Collapse
|
12
|
Yagi C, Kusunoki Y, Tsunoda T, Murase T, Nakamura T, Osugi K, Ohigashi M, Morimoto A, Miyoshi A, Kakutani-Hatayama M, Kosaka-Hamamoto K, Kadoya M, Konishi K, Shoji T, Koyama H. Xanthine oxidoreductase activity is correlated with hepatic steatosis. Sci Rep 2022; 12:12282. [PMID: 35854080 PMCID: PMC9296494 DOI: 10.1038/s41598-022-16688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 07/13/2022] [Indexed: 11/09/2022] Open
Abstract
The enzyme xanthine oxidoreductase (XOR) catalyzes the synthesis of uric acid (UA) from hypoxanthine and xanthine, which are products of purine metabolism starting from ribose-5-phosphate. Several studies suggested a relationship between hyperuricemia and hepatic steatosis; however, few previous studies have directly examined the relationship between XOR activity and hepatic steatosis. A total of 223 subjects with one or more cardiovascular risk factors were enrolled. The liver-to-spleen (L/S) ratio on computed tomography and the hepatic steatosis index (HSI) were used to assess hepatic steatosis. We used a newly developed highly sensitive assay based on [13C2, 15N2] xanthine and liquid chromatography/triple quadrupole mass spectrometry to measure plasma XOR activity. Subjects with the L/S ratio of < 1.1 and the HSI of < 36 had increased XOR activity and serum UA levels. Independent of insulin resistance and serum UA levels, multivariate logistic regression analysis revealed that plasma XOR activity was associated with the risk of hepatic steatosis as assessed by the L/S ratio and HSI. According to the findings of this study, plasma XOR activity is associated with hepatic steatosis independent of insulin resistance and serum UA levels.
Collapse
Affiliation(s)
- Chisako Yagi
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yoshiki Kusunoki
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Taku Tsunoda
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Takayo Murase
- Radioisotope and Chemical Analysis Center, Laboratory Management Department, Sanwa Kagaku Kenkyusho, Nagoya, Japan
| | - Takashi Nakamura
- Radioisotope and Chemical Analysis Center, Laboratory Management Department, Sanwa Kagaku Kenkyusho, Nagoya, Japan
| | - Keiko Osugi
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Mana Ohigashi
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Akiko Morimoto
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Akio Miyoshi
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Miki Kakutani-Hatayama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kae Kosaka-Hamamoto
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Manabu Kadoya
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kosuke Konishi
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Takuhito Shoji
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hidenori Koyama
- Department of Diabetes, Endocrinology and Clinical Immunology, Hyogo Medical University, 1-1, Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| |
Collapse
|
13
|
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
|
14
|
Huang Y, Zhou Y, Wang Y, Chi J, Lv W, Wang Y, Chen Y. The Negative Association Between NAFLD Severity and CKD in a Non-Diabetic Gouty Population. Horm Metab Res 2022; 54:325-334. [PMID: 35378561 DOI: 10.1055/a-1815-7559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD) share common pathogenic mechanisms and risk factors. We aim to evaluate the association between NAFLD and CKD in a non-diabetic gouty population. The retrospective cross sectional study was performed on 1049 non-diabetic gouty participants, who were hospitalized between 2014 and 2020, across 4 districts in Shandong, China. Demographic and clinical characteristics of the study population were collected. The odds ratios (OR) and corresponding 95% confidence intervals (CI) about the NAFLD severity determined by ultrasonography were obtained by multiple logistic regression analysis. An unexpectedly inverse relationship was found between NAFLD severity and the risk of CKD in people with gout. Multivariate logistic regression analysis demonstrated that a higher degree of NAFLD severity is independently associated with a lower risk of CKD in people with gout, after adjusted for age, sex, smoking, gout duration, and metabolic risk factors including obesity, hypertension, hyperglycemia, hyperuricemia, and dyslipidemia, with OR 0.392 (95% CI 0.248-0.619, p<0.001), 0.379 (95% CI 0.233-0.616, p<0.001) and 0.148 (95% CI 0.043-0.512, p=0.003) in participants with mild, moderate, and severe NAFLD, respectively, compared to those without NAFLD. We also observed a weakened association of serum uric acid (SUA) with metabolic risk factors and NAFLD under circumstances of CKD in people with gout (r=-0.054, p=0.466). In conclusion, the presence and severity of NAFLD were negatively associated with the risk of CKD in the non-diabetic gouty population.
Collapse
Affiliation(s)
- Yajing Huang
- Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yue Zhou
- Endocrinology and Metabolism, Fudan University, Shanghai, China
| | - Yahao Wang
- Endocrinology and Metabolism, Fudan University, Shanghai, China
| | - Jingwei Chi
- Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenshan Lv
- Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangang Wang
- Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Chen
- Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Guma M, Dadpey B, Coras R, Mikuls TR, Hamilton B, Quehenberger O, Thorisdottir H, Bittleman D, Lauro K, Reilly SM, Liu-Bryan R, Terkeltaub R. Xanthine oxidase inhibitor urate-lowering therapy titration to target decreases serum free fatty acids in gout and suppresses lipolysis by adipocytes. Arthritis Res Ther 2022; 24:175. [PMID: 35879786 PMCID: PMC9310412 DOI: 10.1186/s13075-022-02852-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/26/2022] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVE Linked metabolic and cardiovascular comorbidities are prevalent in hyperuricemia and gout. For mechanistic insight into impact on inflammatory processes and cardiometabolic risk factors of xanthine oxidase inhibitor urate-lowering therapy (ULT) titration to target, we performed a prospective study of gout serum metabolomes from a ULT trial. METHODS Sera of gout patients meeting the 2015 ACR/EULAR gout classification criteria (n = 20) and with hyperuricemia were studied at time zero and weeks 12 and 24 of febuxostat or allopurinol dose titration ULT. Ultrahigh performance liquid chromatography-tandem mass spectroscopy acquired the serum spectra. Data were assessed using the Metabolon and Metaboloanalyst software. Lipolysis validation assays were done in febuxostat and/or colchicine-treated 3T3-L1 differentiated adipocytes. RESULTS Serum urate decreased from time zero (8.21 ±1.139 SD) at weeks 12 (5.965 ± 1.734 SD) and 24 (5.655 ±1.763 SD). Top metabolites generated by changes in nucleotide and certain amino acid metabolism and polyamine pathways were enriched at 12 and 24 weeks ULT, respectively. Decreases in multiple fatty acid metabolites were observed at 24 weeks, linked with obesity. In cultured adipocytes, febuxostat significantly decreased while colchicine increased the lipolytic response to β-adrenergic-agonism or TNF. CONCLUSION Metabolomic profiles linked xanthine oxidase inhibitor-based ULT titration to target with reduced serum free fatty acids. In vitro validation studies revealed that febuxostat, but not colchicine, reduced lipolysis in cultured adipocytes. Since soluble urate, xanthine oxidase inhibitor treatment, and free fatty acids modulate inflammation, our findings suggest that by suppressing lipolysis, ULT could regulate inflammation in gout and comorbid metabolic and cardiovascular disease.
Collapse
Affiliation(s)
- Monica Guma
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA ,grid.7080.f0000 0001 2296 0625Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona Spain
| | - Benyamin Dadpey
- grid.217200.60000 0004 0627 2787Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093 USA
| | - Roxana Coras
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA ,grid.7080.f0000 0001 2296 0625Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona Spain
| | - Ted R. Mikuls
- grid.266813.80000 0001 0666 4105University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Bartlett Hamilton
- grid.266813.80000 0001 0666 4105University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Oswald Quehenberger
- grid.217200.60000 0004 0627 2787Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093 USA
| | - Hilda Thorisdottir
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| | - David Bittleman
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| | - Kimberly Lauro
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| | - Shannon M. Reilly
- grid.217200.60000 0004 0627 2787Division of Metabolism and Endocrinology, Department of Medicine, University of California-San Diego, La Jolla, CA 92093 USA ,grid.5386.8000000041936877XWeill Center for Metabolic Health, Department of Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Ru Liu-Bryan
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| | - Robert Terkeltaub
- grid.266100.30000 0001 2107 4242Department of Medicine, UC San Diego, San Diego VA Healthcare Service, 3350 La Jolla Village Drive, San Diego, CA 92161 USA
| |
Collapse
|
16
|
Yu S, Li C, Ji G, Zhang L. The Contribution of Dietary Fructose to Non-alcoholic Fatty Liver Disease. Front Pharmacol 2021; 12:783393. [PMID: 34867414 PMCID: PMC8637741 DOI: 10.3389/fphar.2021.783393] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/02/2021] [Indexed: 12/26/2022] Open
Abstract
Fructose, especially industrial fructose (sucrose and high fructose corn syrup) is commonly used in all kinds of beverages and processed foods. Liver is the primary organ for fructose metabolism, recent studies suggest that excessive fructose intake is a driving force in non-alcoholic fatty liver disease (NAFLD). Dietary fructose metabolism begins at the intestine, along with its metabolites, may influence gut barrier and microbiota community, and contribute to increased nutrient absorption and lipogenic substrates overflow to the liver. Overwhelming fructose and the gut microbiota-derived fructose metabolites (e.g., acetate, butyric acid, butyrate and propionate) trigger the de novo lipogenesis in the liver, and result in lipid accumulation and hepatic steatosis. Fructose also reprograms the metabolic phenotype of liver cells (hepatocytes, macrophages, NK cells, etc.), and induces the occurrence of inflammation in the liver. Besides, there is endogenous fructose production that expands the fructose pool. Considering the close association of fructose metabolism and NAFLD, the drug development that focuses on blocking the absorption and metabolism of fructose might be promising strategies for NAFLD. Here we provide a systematic discussion of the underlying mechanisms of dietary fructose in contributing to the development and progression of NAFLD, and suggest the possible targets to prevent the pathogenetic process.
Collapse
Affiliation(s)
- Siyu Yu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunlin Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Lin S, Zhang T, Zhu L, Pang K, Lu S, Liao X, Ying S, Zhu L, Xu X, Wu J, Wang X. Characteristic dysbiosis in gout and the impact of a uric acid-lowering treatment, febuxostat on the gut microbiota. J Genet Genomics 2021; 48:781-791. [PMID: 34509383 DOI: 10.1016/j.jgg.2021.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
Gut dysbiosis is suggested to play a critical role in the pathogenesis of gout. The aim of our study was to identify the characteristic dysbiosis of the gut microbiota in gout patients and the impact of a commonly used uric acid-lowering treatment, febuxostat on gut microbiota in gout. 16S ribosomal RNA sequencing and metagenomic shotgun sequencing was performed on fecal DNA isolated from 38 untreated gout patients, 38 gout patients treated with febuxostat, and 26 healthy controls. A restriction of gut microbiota biodiversity was detected in the untreated gout patients, and the alteration was partly restored by febuxostat. Biochemical metabolic indexes involved in liver and kidney metabolism were significantly associated with the gut microbiota composition in gout patients. Functional analysis revealed that the gut microbiome of gout patients had an enriched function on carbohydrate metabolism but a lower potential for purine metabolism, which was comparatively enhanced in the febuxostat treated gout patients. A classification microbial model obtained a high mean area under the curve up to 0.973. Therefore, gut dysbiosis characterizings gout could potentially serve as a noninvasive diagnostic tool for gout and may be a promising target of future preventive interventions.
Collapse
Affiliation(s)
- Suxian Lin
- Rheumatology Department, Wenzhou People's Hospital, Wenzhou, Zhejiang 325000, China
| | - Tao Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Lingxiao Zhu
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Kun Pang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Saisai Lu
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xin Liao
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Senhong Ying
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Lixia Zhu
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xin Xu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jinyu Wu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xiaobing Wang
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
18
|
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: 3.7] [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
|
19
|
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: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
20
|
Rabaan AA, Tirupathi R, Sule AA, Aldali J, Mutair AA, Alhumaid S, Muzaheed, Gupta N, Koritala T, Adhikari R, Bilal M, Dhawan M, Tiwari R, Mitra S, Emran TB, Dhama K. Viral Dynamics and Real-Time RT-PCR Ct Values Correlation with Disease Severity in COVID-19. Diagnostics (Basel) 2021; 11:1091. [PMID: 34203738 PMCID: PMC8232180 DOI: 10.3390/diagnostics11061091] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 01/08/2023] Open
Abstract
Real-time RT-PCR is considered the gold standard confirmatory test for coronavirus disease 2019 (COVID-19). However, many scientists disagree, and it is essential to understand that several factors and variables can cause a false-negative test. In this context, cycle threshold (Ct) values are being utilized to diagnose or predict SARS-CoV-2 infection. This practice has a significant clinical utility as Ct values can be correlated with the viral load. In addition, Ct values have a strong correlation with multiple haematological and biochemical markers. However, it is essential to consider that Ct values might be affected by pre-analytic, analytic, and post-analytical variables such as collection technique, specimen type, sampling time, viral kinetics, transport and storage conditions, nucleic acid extraction, viral RNA load, primer designing, real-time PCR efficiency, and Ct value determination method. Therefore, understanding the interpretation of Ct values and other influential factors could play a crucial role in interpreting viral load and disease severity. In several clinical studies consisting of small or large sample sizes, several discrepancies exist regarding a significant positive correlation between the Ct value and disease severity in COVID-19. In this context, a revised review of the literature has been conducted to fill the knowledge gaps regarding the correlations between Ct values and severity/fatality rates of patients with COVID-19. Various databases such as PubMed, Science Direct, Medline, Scopus, and Google Scholar were searched up to April 2021 by using keywords including "RT-PCR or viral load", "SARS-CoV-2 and RT-PCR", "Ct value and viral load", "Ct value or COVID-19". Research articles were extracted and selected independently by the authors and included in the present review based on their relevance to the study. The current narrative review explores the correlation of Ct values with mortality, disease progression, severity, and infectivity. We also discuss the factors that can affect these values, such as collection technique, type of swab, sampling method, etc.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia;
| | - Raghavendra Tirupathi
- Department of Medicine Keystone Health, Penn State University School of Medicine, Hershey, PA 16801, USA;
- Department of Medicine, Wellspan Chambersburg and Waynesboro Hospitals, Chambersburg, PA 17201, USA
| | - Anupam A Sule
- Department of Informatics and Outcomes, St Joseph Mercy Oakland, Pontiac, MI 48341, USA;
| | - Jehad Aldali
- Pathology Organization, Imam Mohammed Ibn Saud Islamic University, Riyadh 13317, Saudi Arabia;
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Al-Ahsa 36342, Saudi Arabia;
- College of Nursing, Princess Norah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, NSW 2522, Australia
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Al-Ahsa 31982, Saudi Arabia;
| | - Muzaheed
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Nitin Gupta
- Department of Infectious Diseases, Kasturba Medical College, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Thoyaja Koritala
- Department of Internal Medicine, Mayo Clinic Health System Mankato, Mayo Clinic College of Medicine and Science, Mankato, MN 56001, USA;
| | - Ramesh Adhikari
- Department of Hospital Medicine, Franciscan Health Lafayette, Lafayette, IN 47905, USA;
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, India;
- The Trafford Group of Colleges, Manchester WA14 5PQ, UK
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Prade Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandha Sansthan (DUVASU), Mathura 281001, India;
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India
| |
Collapse
|
21
|
Chang YH, Chiang YF, Chen HY, Huang YJ, Wang KL, Hong YH, Ali M, Shieh TM, Hsia SM. Anti-Inflammatory and Anti-Hyperuricemic Effects of Chrysin on a High Fructose Corn Syrup-Induced Hyperuricemia Rat Model via the Amelioration of Urate Transporters and Inhibition of NLRP3 Inflammasome Signaling Pathway. Antioxidants (Basel) 2021; 10:antiox10040564. [PMID: 33917369 PMCID: PMC8067405 DOI: 10.3390/antiox10040564] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 01/18/2023] Open
Abstract
Hyperuricemia is the main cause of gout and involved in the occurrence of many other diseases such as hyperlipidemia and hypertension correlated with metabolic disorders. Chrysin is a flavonoid compound found naturally in honey, propolis, and mushrooms and has anti-inflammatory and antioxidant effects. However, its mechanism of action is not clear yet. This study investigated the mechanism of chrysin’s anti-hyperuricemic effect in hyperuricemia-induced rats fed with high-fructose corn syrup. Orally administrated chrysin for 28 consecutive days effectively decreased uric acid by inhibiting the activity of xanthine oxidase (XO) in the liver. Moreover, chrysin markedly downregulated the protein expression of uric acid transporter 1 (URAT1) and glucose transporter type 9 (GLUT9) and upregulated the protein expression of organic anion transporter 1 (OAT1) and human ATP-binding cassette subfamily G-2 (ABCG2). In addition, chrysin showed prominent anti-oxidative and inflammatory effects as the malondialdehyde (MDA) and interleukin 1 beta (IL-1β) concentration was reduced in both rat kidney and serum, which aligned with the inhibition of NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome signaling pathway activation. Collectively, our results strongly suggest that chrysin exhibits potent anti-hyperuricemic and anti-inflammatory effects that may yield new adjuvant treatments for gout.
Collapse
Affiliation(s)
- Yi-Hsien Chang
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan;
| | - Yi-Fen Chiang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
| | - Hsin-Yuan Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
- Department of Nutrition, I-Shou University, Kaohsiung 84001, Taiwan;
| | - Yun-Ju Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
| | - Kai-Lee Wang
- Department of Nursing, Ching Kuo Institute of Management and Health, Keelung 20301, Taiwan;
| | - Yong-Han Hong
- Department of Nutrition, I-Shou University, Kaohsiung 84001, Taiwan;
| | - Mohamed Ali
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt;
| | - Tzong-Ming Shieh
- School of Dentistry, College of Dentistry, China Medical University, Taichung 40402, Taiwan;
| | - Shih-Min Hsia
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan;
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; (Y.-F.C.); (H.-Y.C.); (Y.-J.H.)
- School of Food and Safety, Taipei Medical University, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Correspondence: ; Tel.: +886-273-61661-6558
| |
Collapse
|
22
|
Andreadou I, Daiber A, Baxter GF, Brizzi MF, Di Lisa F, Kaludercic N, Lazou A, Varga ZV, Zuurbier CJ, Schulz R, Ferdinandy P. Influence of cardiometabolic comorbidities on myocardial function, infarction, and cardioprotection: Role of cardiac redox signaling. Free Radic Biol Med 2021; 166:33-52. [PMID: 33588049 DOI: 10.1016/j.freeradbiomed.2021.02.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 02/06/2023]
Abstract
The morbidity and mortality from cardiovascular diseases (CVD) remain high. Metabolic diseases such as obesity, hyperlipidemia, diabetes mellitus (DM), non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) as well as hypertension are the most common comorbidities in patients with CVD. These comorbidities result in increased myocardial oxidative stress, mainly from increased activity of nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, mitochondria as well as downregulation of antioxidant defense systems. Oxidative and nitrosative stress play an important role in ischemia/reperfusion injury and may account for increased susceptibility of the myocardium to infarction and myocardial dysfunction in the presence of the comorbidities. Thus, while early reperfusion represents the most favorable therapeutic strategy to prevent ischemia/reperfusion injury, redox therapeutic strategies may provide additive benefits, especially in patients with heart failure. While oxidative and nitrosative stress are harmful, controlled release of reactive oxygen species is however important for cardioprotective signaling. In this review we summarize the current data on the effect of hypertension and major cardiometabolic comorbidities such as obesity, hyperlipidemia, DM, NAFLD/NASH on cardiac redox homeostasis as well as on ischemia/reperfusion injury and cardioprotection. We also review and discuss the therapeutic interventions that may restore the redox imbalance in the diseased myocardium in the presence of these comorbidities.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece.
| | - Andreas Daiber
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr, Germany.
| | - Gary F Baxter
- Division of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, United Kingdom
| | | | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Italy; Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany.
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
23
|
Yang Y, Zhang JL, Zhou Q. Targets and mechanisms of dietary anthocyanins to combat hyperglycemia and hyperuricemia: a comprehensive review. Crit Rev Food Sci Nutr 2020; 62:1119-1143. [PMID: 33078617 DOI: 10.1080/10408398.2020.1835819] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hyperglycemia and hyperuricemia are both metabolic disorders related to excessive amount of metabolites in blood, which are considered as high risk factors for the development of many chronic diseases. Enzymes, cells, tissues and organs, which are relevant to metabolism and excretion of glucose and UA, are usually regarded to be the targets in treatment of hyperglycemia and hyperuricemia. Several drugs have been commonly applied to combat hyperglycemia and hyperuricemia through various targets but with unignorable side effects. Anthocyanins have become promising alternatives against hyperglycemia and hyperuricemia because of their bio-activities with little side effects. Structurally different anthocyanins from berry fruits, cherries and purple sweet potato lead to the diverse functional activity and property. This review is aimed to illustrate the specific targets that are available for anthocyanins from berry fruits, cherries and purple sweet potato in hyperglycemia and hyperuricemia management, as well as discuss the structure-activity relationship, and the underlying mechanisms associated with intracellular signaling pathway, anti-oxidative stress and anti-inflammation. In addition, the relationship of hyperglycemia and hyperuricemia, and the possibly regulative role of anthocyanins against them, along with the effects of anthocyanins in clinical trial are mentioned.
Collapse
Affiliation(s)
- Yang Yang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jiu-Liang Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China.,Ministry of Education, Key Laboratory of Environment Correlative Dietology, Wuhan, China
| | - Qing Zhou
- Department of Pharmacy, Wuhan City Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
High fat diet-triggered non-alcoholic fatty liver disease: A review of proposed mechanisms. Chem Biol Interact 2020; 330:109199. [DOI: 10.1016/j.cbi.2020.109199] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
|
25
|
Russo E, Leoncini G, Esposito P, Garibotto G, Pontremoli R, Viazzi F. Fructose and Uric Acid: Major Mediators of Cardiovascular Disease Risk Starting at Pediatric Age. Int J Mol Sci 2020; 21:E4479. [PMID: 32599713 PMCID: PMC7352635 DOI: 10.3390/ijms21124479] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Recently, there has been a growing interest in epidemiological and clinical studies supporting a pathogenetic role of fructose in cardio-metabolic diseases, especially in children and adolescents. In the present review, we summarize experimental data on the potential biological mechanisms linking fructose and uric acid in the development of insulin resistance, metabolic syndrome, obesity, diabetes, hypertension, non-alcoholic fatty liver disease and chronic renal disease, thereby contributing to an increase in cardiovascular risk at pediatric age.
Collapse
Affiliation(s)
- Elisa Russo
- Clinica Nefrologica, Ospedale Policlinico San Martino, Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, 16132 Genova, Italy; (E.R.); (P.E.); (G.G.)
| | - Giovanna Leoncini
- Clinica di Medicina Interna 2, Ospedale Policlinico San Martino, Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, 16132 Genova, Italy; (G.L.); (R.P.)
| | - Pasquale Esposito
- Clinica Nefrologica, Ospedale Policlinico San Martino, Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, 16132 Genova, Italy; (E.R.); (P.E.); (G.G.)
| | - Giacomo Garibotto
- Clinica Nefrologica, Ospedale Policlinico San Martino, Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, 16132 Genova, Italy; (E.R.); (P.E.); (G.G.)
| | - Roberto Pontremoli
- Clinica di Medicina Interna 2, Ospedale Policlinico San Martino, Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, 16132 Genova, Italy; (G.L.); (R.P.)
| | - Francesca Viazzi
- Clinica Nefrologica, Ospedale Policlinico San Martino, Dipartimento di Medicina Interna e Specialità Mediche, Università degli Studi di Genova, 16132 Genova, Italy; (E.R.); (P.E.); (G.G.)
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW This article reviews evidence linking cardiometabolic conditions with changes in purine metabolites, including increased serum uric acid (sUA), and discusses intervention studies that investigated the therapeutic relevance of these associations. RECENT FINDINGS Metabolic and epidemiological findings support a correlation between sUA and circulating levels of other purines with insulin resistance (IR) and risk factors for cardiovascular disease (CVD). In addition, increased activity of xanthine oxidoreductase (XOR), the rate-limiting enzyme for UA production, has been detected in tissues targeted by obesity. Yet, inhibition of XOR in pre-clinical and clinical studies generally failed to support a causal role for excess sUA in IR and CVD. The lack of efficacy of XOR inhibitors strongly suggests that UA is a marker of, rather than a direct contributory factor for, cardiometabolic diseases. Validation of the function of other purines will require a paradigm shift, from a "UA-centric" view to a more granular assessment of the entire purine network and its interaction with other pathways.
Collapse
Affiliation(s)
- Giulio R Romeo
- Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA.
| | - Meenu Jain
- Lahey Hospital & Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| |
Collapse
|
27
|
Brennan P, Clare K, George J, Dillon JF. Determining the role for uric acid in non-alcoholic steatohepatitis development and the utility of urate metabolites in diagnosis: An opinion review. World J Gastroenterol 2020; 26:1683-1690. [PMID: 32351286 PMCID: PMC7183862 DOI: 10.3748/wjg.v26.i15.1683] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/02/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
There has long been a recognised association between non-alcoholic fatty liver disease (NAFLD) and the composite aspects of the metabolic syndrome. Part of this association highlighted the supposed co-existence of elevated uric acid levels in those with NAFLD. There is interest in exploitation of this as a putative diagnostic and prognostic biomarker in NAFLD. Given the increased economic and health burden associated with the NAFLD epidemic, improved methods of population-based, minimally-invasive methods and biomarkers are clearly highly sought and necessary. In this opinion review we review the proposed role of uric acid in the pathogenesis of NAFLD and its potential utilisation in the diagnosis and monitoring of the disease process.
Collapse
Affiliation(s)
- Paul Brennan
- Department of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| | - Kathleen Clare
- Department of Neurology, Queen Elizabeth Hospital, Glasgow G51 4TF, United Kingdom
| | - Jacob George
- Department of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| | - John F Dillon
- Department of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| |
Collapse
|
28
|
Lu J, Dalbeth N, Yin H, Li C, Merriman TR, Wei WH. Mouse models for human hyperuricaemia: a critical review. Nat Rev Rheumatol 2020; 15:413-426. [PMID: 31118497 DOI: 10.1038/s41584-019-0222-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hyperuricaemia (increased serum urate concentration) occurs mainly in higher primates, including in humans, because of inactivation of the gene encoding uricase during primate evolution. Individuals with hyperuricaemia might develop gout - a painful inflammatory arthritis caused by monosodium urate crystal deposition in articular structures. Hyperuricaemia is also associated with common chronic diseases, including hypertension, chronic kidney disease, type 2 diabetes and cardiovascular disease. Many mouse models have been developed to investigate the causal mechanisms for hyperuricaemia. These models are highly diverse and can be divided into two broad categories: mice with genetic modifications (genetically induced models) and mice exposed to certain environmental factors (environmentally induced models; for example, pharmaceutical or dietary induction). This Review provides an overview of the mouse models of hyperuricaemia and the relevance of these models to human hyperuricaemia, with an emphasis on those models generated through genetic modifications. The challenges in developing and comparing mouse models of hyperuricaemia and future research directions are also outlined.
Collapse
Affiliation(s)
- Jie Lu
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand.,Shandong Provincial Key Laboratory of Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, the Affiliated Hospital of Qingdao University, Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Huiyong Yin
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), CAS, Shanghai, China
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, the Affiliated Hospital of Qingdao University, Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand.
| | - Wen-Hua Wei
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
29
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
30
|
Xanthine Oxidase Inhibitor Febuxostat Exerts an Anti-Inflammatory Action and Protects against Diabetic Nephropathy Development in KK-Ay Obese Diabetic Mice. Int J Mol Sci 2019; 20:ijms20194680. [PMID: 31546603 PMCID: PMC6801943 DOI: 10.3390/ijms20194680] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/14/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022] Open
Abstract
Hyperuricemia has been recognized as a risk factor for insulin resistance as well as one of the factors leading to diabetic kidney disease (DKD). Since DKD is the most common cause of end-stage renal disease, we investigated whether febuxostat, a xanthine oxidase (XO) inhibitor, exerts a protective effect against the development of DKD. We used KK-Ay mice, an established obese diabetic rodent model. Eight-week-old KK-Ay mice were provided drinking water with or without febuxostat (15 μg/mL) for 12 weeks and then subjected to experimentation. Urine albumin secretion and degrees of glomerular injury judged by microscopic observations were markedly higher in KK-Ay than in control lean mice. These elevations were significantly normalized by febuxostat treatment. On the other hand, body weights and high serum glucose concentrations and glycated albumin levels of KK-Ay mice were not affected by febuxostat treatment, despite glucose tolerance and insulin tolerance tests having revealed febuxostat significantly improved insulin sensitivity and glucose tolerance. Interestingly, the IL-1β, IL-6, MCP-1, and ICAM-1 mRNA levels, which were increased in KK-Ay mouse kidneys as compared with normal controls, were suppressed by febuxostat administration. These data indicate a protective effect of XO inhibitors against the development of DKD, and the underlying mechanism likely involves inflammation suppression which is independent of hyperglycemia amelioration.
Collapse
|
31
|
Oral A, Sahin T, Turker F, Kocak E. Relationship Between Serum Uric Acid Levels and Nonalcoholic Fatty Liver Disease in Non-Obese Patients. ACTA ACUST UNITED AC 2019; 55:medicina55090600. [PMID: 31533345 PMCID: PMC6780528 DOI: 10.3390/medicina55090600] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/09/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023]
Abstract
Background and objectives: Nonalcoholic fatty liver disease (NAFLD) is associated with multiple factors such as hypertension, diabetes, dyslipidemia, obesity, and hyperuricemia. We aim to investigate the relationship between uric acid and NAFLD in a non-obese and young population. Materials and Methods: This study was performed in January 2010–2019 with a group of 367 (225 patients in the NAFLD group and 142 in the control group) patients with liver biopsy-proven NAFLD or no NAFLD. Patients with NAFLD were classified according to the percentage of steatosis as follows, group I had 1–20% and group II >20%. Demographic, clinical, and laboratory (biochemical parameters) features were collected retrospectively. Results: The mean body mass index (BMI) and age of the patients were 26.41 ± 3.42 and 32.27 ± 8.85, respectively. The BMI, homeostatic model of assessment (HOMA-IR), and uric acid (UA) values of the NAFLD group were found to be significantly higher than those of the controls. A positive correlation was found between the NAFLD stage and UA. The following factors were independently associated with NAFLD: BMI, HOMA-IR, and UA. In addition, the cut-off value of UA was 4.75 mg/dl with a sensitivity of 45.8% and a specificity of 80.3%. Conclusions: UA is a simple, non-invasive, cheap, and useful marker that may be used to predict steatosis in patients with NAFLD.
Collapse
Affiliation(s)
- Alihan Oral
- Department of Internal Medicine, Faculty of Medicine, Demiroglu Bilim University, 34360 Istanbul, Turkey.
| | - Tolga Sahin
- Department of Gastroenterology, Faculty of Medicine, Demiroglu Bilim University, 34360 Istanbul, Turkey.
| | - Fatih Turker
- Department of Internal Medicine, Faculty of Medicine, Demiroglu Bilim University, 34360 Istanbul, Turkey.
| | - Erdem Kocak
- Department of Gastroenterology, Faculty of Medicine, Demiroglu Bilim University, 34360 Istanbul, Turkey.
| |
Collapse
|
32
|
Heikal MM, Shaaban AA, Elkashef WF, Ibrahim TM. Effect of febuxostat on biochemical parameters of hyperlipidemia induced by a high-fat diet in rabbits. Can J Physiol Pharmacol 2019; 97:611-622. [DOI: 10.1139/cjpp-2018-0731] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Febuxostat, a highly potent xanthine oxidase inhibitor with an antioxidant effect, inhibits elevated xanthine oxidase, leading to reduction of reactive oxygen species and oxidative stress, the main causes of vascular inflammation in hyperlipidemia. The aim of this study was to test the potential antioxidant and anti-inflammatory effects of febuxostat and (or) stopping a high-fat diet on the biochemical parameters in rabbits with hyperlipidemia induced by a high-fat diet. Male New Zealand rabbits were distributed into 3 groups: a normal control group fed standard chow for 12 weeks and 2 other groups fed a high-fat diet with 1% cholesterol for 8 weeks, and then shifted to standard chow for 4 weeks. During the last 4 weeks, one high-fat diet group received 0.5% carboxymethyl cellulose, whereas the other group was treated with febuxostat (2 mg/kg per day p.o.). Febuxostat significantly lowered low-density lipoprotein cholesterol (“bad” cholesterol) compared to the untreated group (high-fat diet group). Febuxostat also displayed a potent anti-inflammatory and antioxidant activity by decreasing serum levels of lipid peroxidation index, proinflammatory cytokines, and enhancing antioxidant enzyme activity. Stopping the hyperlipidemic diet in the high-fat diet group did not show improvement. These findings indicate the antioxidant and anti-inflammatory effects of febuxostat that may be common mechanisms of the anti-hyperlipidemic effect of this drug. Stopping a hyperlipidemic diet without treatment is not sufficient once injury has occurred.
Collapse
Affiliation(s)
- Mohammed M. Heikal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed A. Shaaban
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Faculty of Pharmacy, Aqaba University of Technology, Jordan
| | - Wagdi F. Elkashef
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Tarek M. Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
33
|
Golonka R, Yeoh BS, Vijay-Kumar M. Dietary Additives and Supplements Revisited: The Fewer, the Safer for Liver and Gut Health. ACTA ACUST UNITED AC 2019; 5:303-316. [PMID: 32864300 DOI: 10.1007/s40495-019-00187-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose of Review The supplementation of dietary additives into processed foods has exponentially increased in the past few decades. Similarly, the incidence rates of various diseases, including metabolic syndrome, gut dysbiosis and hepatocarcinogenesis, have been elevating. Current research reveals that there is a positive association between food additives and these pathophysiological diseases. This review highlights the research published within the past 5 years that elucidate and update the effects of dietary supplements on liver and intestinal health. Recent Findings Some of the key findings include: enterocyte dysfunction of fructose clearance causes non-alcoholic fatty liver disease (NAFLD); non-caloric sweeteners are hepatotoxic; dietary emulsifiers instigate gut dysbiosis and hepatocarcinogenesis; and certain prebiotics can induce cholestatic hepatocellular carcinoma (HCC) in gut dysbiotic mice. Overall, multiple reports suggest that the administration of purified, dietary supplements could cause functional damage to both the liver and gut. Summary The extraction of bioactive components from natural resources was considered a brilliant method to modulate human health. However, current research highlights that such purified components may negatively affect individuals with microbiotal dysbiosis, resulting in a deeper break of the symbiotic relationship between the host and gut microbiota, which can lead to repercussions on gut and liver health. Therefore, ingestion of these dietary additives should not go without some caution!
Collapse
Affiliation(s)
- Rachel Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- Graduate Program in Immunology & Infectious Disease, Pennsylvania State University, University Park, PA 16802, USA
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA.,Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
34
|
Harmon DB, Mandler WK, Sipula IJ, Dedousis N, Lewis SE, Eckels JT, Du J, Wang Y, Huckestein BR, Pagano PJ, Cifuentes-Pagano E, Homanics GE, Van't Erve TJ, Stefanovic-Racic M, Jurczak MJ, O'Doherty RM, Kelley EE. Hepatocyte-Specific Ablation or Whole-Body Inhibition of Xanthine Oxidoreductase in Mice Corrects Obesity-Induced Systemic Hyperuricemia Without Improving Metabolic Abnormalities. Diabetes 2019; 68:1221-1229. [PMID: 30936145 PMCID: PMC6610025 DOI: 10.2337/db18-1198] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/18/2019] [Indexed: 01/20/2023]
Abstract
Systemic hyperuricemia (HyUA) in obesity/type 2 diabetes facilitated by elevated activity of xanthine oxidoreductase (XOR), which is the sole source of uric acid (UA) in mammals, has been proposed to contribute to the pathogenesis of insulin resistance/dyslipidemia in obesity. Here, the effects of hepatocyte-specific ablation of Xdh, the gene encoding XOR (HXO), and whole-body pharmacologic inhibition of XOR (febuxostat) on obesity-induced insulin resistance/dyslipidemia were assessed. Deletion of hepatocyte Xdh substantially lowered liver and plasma UA concentration. When exposed to an obesogenic diet, HXO and control floxed (FLX) mice became equally obese, but systemic HyUA was absent in HXO mice. Despite this, obese HXO mice became as insulin resistant and dyslipidemic as obese FLX mice. Similarly, febuxostat dramatically lowered plasma and tissue UA and XOR activity in obese wild-type mice without altering obesity-associated insulin resistance/dyslipidemia. These data demonstrate that hepatocyte XOR activity is a critical determinant of systemic UA homeostasis, that deletion of hepatocyte Xdh is sufficient to prevent systemic HyUA of obesity, and that neither prevention nor correction of HyUA improves insulin resistance/dyslipidemia in obesity. Thus, systemic HyUA, although clearly a biomarker of the metabolic abnormalities of obesity, does not appear to be causative.
Collapse
Affiliation(s)
- Daniel B Harmon
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA
| | - W Kyle Mandler
- Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV
| | - Ian J Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Nikolaos Dedousis
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Sara E Lewis
- Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV
| | - Jeremy T Eckels
- Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV
| | - Jianhai Du
- Department of Ophthalmology and Biochemistry, Health Sciences Center, West Virginia University, Morgantown, WV
| | - Yekai Wang
- Department of Ophthalmology and Biochemistry, Health Sciences Center, West Virginia University, Morgantown, WV
| | - Brydie R Huckestein
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Patrick J Pagano
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA
| | - Eugenia Cifuentes-Pagano
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA
| | - Gregg E Homanics
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute and Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Thomas J Van't Erve
- Immunity, Inflammation, and Disease Laboratory/Free Radical Metabolism Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC
| | - Maja Stefanovic-Racic
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Robert M O'Doherty
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Eric E Kelley
- Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV
| |
Collapse
|
35
|
Yeo YH, Lai YC. Redox Regulation of Metabolic Syndrome: Recent Developments in Skeletal Muscle Insulin Resistance and Non-alcoholic Fatty Liver Disease (NAFLD). CURRENT OPINION IN PHYSIOLOGY 2019; 9:79-86. [PMID: 32818162 DOI: 10.1016/j.cophys.2019.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Several new discoveries over the past decade have shown that metabolic syndrome, a cluster of metabolic disorders, including increased visceral obesity, hyperglycemia, hypertension, dyslipidemia and low HDL-cholesterol, is commonly associated with skeletal muscle insulin resistance. More recently, non-alcoholic fatty liver disease (NAFLD) was recognized as an additional condition that is strongly associated with features of metabolic syndrome. While the pathogenesis of skeletal muscle insulin resistance and fatty liver is multifactorial, the role of dysregulated redox signaling has been clearly demonstrated in the regulation of skeletal muscle insulin resistance and NAFLD. In this review, we aim to provide recent updates on redox regulation with respect to (a) pro-oxidant enzymes (e.g. NAPDH oxidase and xanthine oxidase); (b) mitochondrial dysfunction; (c) endoplasmic reticulum (ER) stress; (d) iron metabolism derangements; and (e) gut-skeletal muscle or gut-liver connection in the development of skeletal muscle insulin resistance and NAFLD. Furthermore, we discuss promising new therapeutic strategies targeting redox regulation currently under investigation for the treatment of skeletal muscle insulin resistance and NAFLD.
Collapse
Affiliation(s)
- Yee-Hui Yeo
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, Palo Alto, California, USA
| | - Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine; Indianapolis, IN, USA
| |
Collapse
|
36
|
Sanchez-Lozada LG, Andres-Hernando A, Garcia-Arroyo FE, Cicerchi C, Li N, Kuwabara M, Roncal-Jimenez CA, Johnson RJ, Lanaspa MA. Uric acid activates aldose reductase and the polyol pathway for endogenous fructose and fat production causing development of fatty liver in rats. J Biol Chem 2019; 294:4272-4281. [PMID: 30651350 DOI: 10.1074/jbc.ra118.006158] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/10/2019] [Indexed: 12/18/2022] Open
Abstract
Dietary, fructose-containing sugars have been strongly associated with the development of nonalcoholic fatty liver disease (NAFLD). Recent studies suggest that fructose also can be produced via the polyol pathway in the liver, where it may induce hepatic fat accumulation. Moreover, fructose metabolism yields uric acid, which is highly associated with NAFLD. Here, using biochemical assays, reporter gene expression, and confocal fluorescence microscopy, we investigated whether uric acid regulates aldose reductase, a key enzyme in the polyol pathway. We evaluated whether soluble uric acid regulates aldose reductase expression both in cultured hepatocytes (HepG2 cells) and in the liver of hyperuricemic rats and whether this stimulation is associated with endogenous fructose production and fat accumulation. Uric acid dose-dependently stimulated aldose reductase expression in the HepG2 cells, and this stimulation was associated with endogenous fructose production and triglyceride accumulation. This stimulatory mechanism was mediated by uric acid-induced oxidative stress and stimulation of the transcription factor nuclear factor of activated T cells 5 (NFAT5). Uric acid also amplified the effects of elevated glucose levels to stimulate hepatocyte triglyceride accumulation. Hyperuricemic rats exhibited elevated hepatic aldose reductase expression, endogenous fructose accumulation, and fat buildup that was significantly reduced by co-administration of the xanthine oxidase inhibitor allopurinol. These results suggest that uric acid generated during fructose metabolism may act as a positive feedback mechanism that stimulates endogenous fructose production by stimulating aldose reductase in the polyol pathway. Our findings suggest an amplifying mechanism whereby soft drinks rich in glucose and fructose can induce NAFLD.
Collapse
Affiliation(s)
- Laura G Sanchez-Lozada
- From the Laboratory of Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, CP 14080 Mexico City, Mexico and
| | - Ana Andres-Hernando
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Fernando E Garcia-Arroyo
- From the Laboratory of Renal Physiopathology, Instituto Nacional de Cardiología Ignacio Chavez, CP 14080 Mexico City, Mexico and
| | - Christina Cicerchi
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Nanxing Li
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Masanari Kuwabara
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Carlos A Roncal-Jimenez
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Richard J Johnson
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| | - Miguel A Lanaspa
- the Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado, Aurora, Colorado 80045
| |
Collapse
|
37
|
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: 1.5] [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
|
38
|
Wang Z, Wang X, Yan H, Liu Y, Li L, Li S, Wang X, Wang D. Aronia melanocarpa ameliorates gout and hyperuricemia in animal models. FOOD AGR IMMUNOL 2018. [DOI: 10.1080/09540105.2018.1541967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Zhuqian Wang
- School of Life Sciences, Jilin University, Changchun, People’s Republic of China
| | - Xi Wang
- School of Life Sciences, Jilin University, Changchun, People’s Republic of China
| | - Han Yan
- School of Life Sciences, Jilin University, Changchun, People’s Republic of China
| | - Yan Liu
- School of Life Sciences, Jilin University, Changchun, People’s Republic of China
| | - Lanzhou Li
- School of Life Sciences, Jilin University, Changchun, People’s Republic of China
| | - Shaopeng Li
- School of Pharmacy and Food Science, Zhuhai College of Jilin University, Jilin University, Zhuhai, People’s Republic of China
| | - Xiaofeng Wang
- Department of Stomatology, China–Japan Union Hospital of Jilin University, Jilin University, Changchun, People’s Republic of China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, People’s Republic of China
- School of Pharmacy and Food Science, Zhuhai College of Jilin University, Jilin University, Zhuhai, People’s Republic of China
| |
Collapse
|
39
|
Dangana EO, Michael OS, Omolekulo TE, Areola ED, Olatunji LA. Enhanced hepatic glycogen synthesis and suppressed adenosine deaminase activity by lithium attenuates hepatic triglyceride accumulation in nicotine-exposed rats. Biomed Pharmacother 2018; 109:1417-1427. [PMID: 30551393 DOI: 10.1016/j.biopha.2018.10.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/05/2018] [Accepted: 10/12/2018] [Indexed: 01/08/2023] Open
Abstract
Reduced liver glycogen synthesis might signify increased glucose flux towards fat synthesis and triggers hepatic triglyceride accumulation and dysmetabolism. Adenosine deaminase (ADA) reduces adenosine content which increases glycogenolysis. In the present study, we evaluate the effect of modulating glycogen synthesis and ADA by lithium chloride (LiCl) on nicotine-induced dysmetabolism. Twenty four male Wistar rats (n = 6/group) were allotted into four groups namely; vehicle-treated (po), nicotine-treated (1.0 mg/kg; po), LiCl-treated (5.0 mg/kg; po) and nicotine + LiCl-treated groups. The treatments lasted for 8 weeks. Nicotine exposure resulted in reduced body weight gain, liver weight, visceral adiposity, glycogen content and synthase. Along with increased insulin resistance (IR), fasting plasma glucose, lactate, plasma and hepatic ADA, XO, UA, and triglyceride (TG), total cholesterol (TC), free fatty acid, lipid peroxidation and liver injury markers. However, plasma and hepatic glucose-6-phosphate dehydrogenase-dependent antioxidant defenses were not affected by nicotine exposure. Concurrent treatment with LiCl normalizes all alterations with exception of hepatic TC. This result shows that enhancement of hepatic glycogen synthesis and suppression of ADA/XO/uric acid pathway by lithium can salvage the liver from nicotine-induced TG accumulation.
Collapse
Affiliation(s)
- Elizabeth O Dangana
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olugbenga S Michael
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria; Cardiometabolic Research Unit, Department of Physiology, College of Health Sciences, Bowen University Iwo, Nigeria
| | - Tolulope E Omolekulo
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Emmanuel D Areola
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A Olatunji
- HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.
| |
Collapse
|
40
|
Sodium acetate improves disrupted glucoregulation and hepatic triglyceride content in insulin-resistant female rats: involvement of adenosine deaminase and dipeptidyl peptidase-4 activities. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:103-116. [PMID: 30280312 DOI: 10.1007/s00210-018-1569-2] [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] [Received: 07/06/2018] [Accepted: 09/25/2018] [Indexed: 12/15/2022]
Abstract
Combined oral contraceptive (COC) treatment has been shown to be associated with glucose deregulation and increased triglyceride levels, but the mechanisms are elusive. Soluble dipeptidyl peptidase-4 (sDPP-4) and adenosine deaminase (ADA) are involved in the initiation and/or progression of cardiometabolic disorders. We therefore, hypothesized that increased DPP-4 and ADA activities are involved in glucose deregulation and hepatic triglyceride accumulation induced by COC treatment. This study also investigated whether short-chain fatty acid, acetate, would protect against COC-induced dysmetabolic effects. Female Wistar rats received (p.o.) vehicle and COC (1.0 μg ethinylestradiol plus 5.0 μg levonorgestrel) with or without sodium acetate (ACE; 200 mg) for 8 weeks. Treatment with COC led to increased plasma triglyceride-glucose index, 1-h postload glucose response, insulin, free fatty acid, insulin resistance, and impaired glucose tolerance. COC treatment also resulted in increased plasma and hepatic triglycerides (TG), TG/HDL-cholesterol ratio, malondialdehyde, uric acid, lactate dehydrogenase, DPP-4, ADA, and xanthine oxidase (XO) activities. On the other hand, COC led to reduction in nitric oxide level. However, ACE significantly ameliorated the alterations induced by COC treatment, but XO activity remains elevated during COC treatment. This result also demonstrates that increased DPP-4 and ADA activities are at least in part involved in glucose deregulation and hepatic TG accumulation induced by COC treatment. Therefore, sodium acetate would impact positively on cardiometabolic disorders, at least in part, by inhibition of DPP-4 and ADA activities.
Collapse
|
41
|
Increased Serum Uric Acid over five years is a Risk Factor for Developing Fatty Liver. Sci Rep 2018; 8:11735. [PMID: 30082907 PMCID: PMC6079004 DOI: 10.1038/s41598-018-30267-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022] Open
Abstract
The prevalence of fatty liver disease (FLD) is increasing. To clarify risk factors for developing FLD, we analyzed a database from healthy Japanese adults who had annual medical check-ups in 2004 and reexamined in 2009. We used the fatty liver index (FLI) to classify participants as FLD (FLI ≥60), borderline FLD (30≤ FLI <60), and normal liver (FLI <30). Subjects with hepatitis B or C virus infection and subjects with FLD at the baseline were excluded. The cumulative incidence of FLD from normal liver and from borderline FLD over five years were 0.65% (52/8,025) and 12.9% (244/1,888), respectively. After multiple adjustments, higher serum uric acid (SUA) (OR:1.92; 95% CI:1.40–2.63) and increased SUA change (OR:3.734; 95% CI:2.57–5.42) became risk factors for developing FLD from normal liver, as well as younger age and higher body mass index. The risk factors for developing FLD from borderline FLD were similar. Not only higher baseline SUA but also increased SUA change became independent risks for developing FLD.
Collapse
|
42
|
Jensen T, Abdelmalek MF, Sullivan S, Nadeau KJ, Green M, Roncal C, Nakagawa T, Kuwabara M, Sato Y, Kang DH, Tolan DR, Sanchez-Lozada LG, Rosen HR, Lanaspa MA, Diehl AM, Johnson RJ. Fructose and sugar: A major mediator of non-alcoholic fatty liver disease. J Hepatol 2018; 68:1063-1075. [PMID: 29408694 PMCID: PMC5893377 DOI: 10.1016/j.jhep.2018.01.019] [Citation(s) in RCA: 536] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 12/11/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome; its rising prevalence parallels the rise in obesity and diabetes. Historically thought to result from overnutrition and a sedentary lifestyle, recent evidence suggests that diets high in sugar (from sucrose and/or high-fructose corn syrup [HFCS]) not only increase the risk of NAFLD, but also non-alcoholic steatohepatitis (NASH). Herein, we review the experimental and clinical evidence that fructose precipitates fat accumulation in the liver, due to both increased lipogenesis and impaired fat oxidation. Recent evidence suggests that the predisposition to fatty liver is linked to the metabolism of fructose by fructokinase C, which results in ATP consumption, nucleotide turnover and uric acid generation that mediate fat accumulation. Alterations to gut permeability, the microbiome, and associated endotoxemia contribute to the risk of NAFLD and NASH. Early clinical studies suggest that reducing sugary beverages and total fructose intake, especially from added sugars, may have a significant benefit on reducing hepatic fat accumulation. We suggest larger, more definitive trials to determine if lowering sugar/HFCS intake, and/or blocking uric acid generation, may help reduce NAFLD and its downstream complications of cirrhosis and chronic liver disease.
Collapse
Affiliation(s)
- Thomas Jensen
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| | | | - Shelby Sullivan
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kristen J Nadeau
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Melanie Green
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Carlos Roncal
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Takahiko Nakagawa
- Division of Future Basic Medicine, Nara Medical University, Nara, Japan
| | - Masanari Kuwabara
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Yuka Sato
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Duk-Hee Kang
- Division of Nephrology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Dean R Tolan
- Dept of Biology, Boston University, Boston, MA, United States
| | | | - Hugo R Rosen
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Miguel A Lanaspa
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | | | - Richard J Johnson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
43
|
Kabil SL, Mahmoud NM. Canagliflozin protects against non-alcoholic steatohepatitis in type-2 diabetic rats through zinc alpha-2 glycoprotein up-regulation. Eur J Pharmacol 2018; 828:135-145. [PMID: 29608898 DOI: 10.1016/j.ejphar.2018.03.043] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023]
Abstract
Elevated blood glucose and insulin resistance are triggering factors for non-alcoholic steatohepatitis (NASH). We investigated the effects of the Sodium Glucose co-Transporter 2 (SGLT2) inhibitor canagliflozin on NASH development in rats with type 2 diabetes mellitus as well as the possible underlying mechanisms and for the first time the effect of canagliflozin on the hepatic zinc-α2-glycoprotein (ZAG) levels. Rats were treated with nicotinamide and streptozotocin to reduce the insulin secretory capacity then fed high fat diet for 8 weeks. The diabetic high fat diet rats were divided into three groups; untreated group, canagliflozin 10 mg/kg treated group and canagliflozin 20 mg/kg treated group during this period. The elevated blood glucose and glycated haemoglobin (HbA1c) levels in the diabetic high fat diet rats were significantly reduced by canagliflozin. Moreover, the diabetic high fat diet induced NASH development as evidenced by liver weight gain, hepatic lipid accumulation and low hepatic ZAG expression as well as increased serum alanine aminotransferase; all these changes were reversed in rats treated with canagliflozin. Additionally, canagliflozin succeeded to upregulate the hepatic ZAG levels in both normal and diabetic high fat fed rats, lower the serum and hepatic inflammatory cytokines levels as well as lower the serum caspase-3 levels and enhanced hepatic Bcl-2 expression. Also, canagliflozin attenuated hepatic oxidative stress and elevated the antioxidant enzymes activity as well as the total antioxidant capacity. All these effects of canagliflozin were dose dependant. CONCLUSION SGLT2 inhibitor-canagliflozin- has beneficial effects in treatment of NASH associated with diabetes mellitus.
Collapse
Affiliation(s)
- Soad L Kabil
- Department of Pharmacology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Nevertyty M Mahmoud
- Department of Pharmacology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
44
|
Mucke HA. Drug Repurposing Patent Applications April–June 2017. Assay Drug Dev Technol 2017; 15:372-377. [DOI: 10.1089/adt.2017.29068.pq2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
45
|
Lonardo A, Nascimbeni F, Maurantonio M, Marrazzo A, Rinaldi L, Adinolfi LE. Nonalcoholic fatty liver disease: Evolving paradigms. World J Gastroenterol 2017; 23:6571-6592. [PMID: 29085206 PMCID: PMC5643282 DOI: 10.3748/wjg.v23.i36.6571] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/21/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023] Open
Abstract
In the last years new evidence has accumulated on nonalcoholic fatty liver disease (NAFLD) challenging the paradigms that had been holding the scene over the previous 30 years. NAFLD has such an epidemic prevalence as to make it impossible to screen general population looking for NAFLD cases. Conversely, focusing on those cohorts of individuals exposed to the highest risk of NAFLD could be a more rational approach. NAFLD, which can be diagnosed with either non-invasive strategies or through liver biopsy, is a pathogenically complex and clinically heterogeneous disease. The existence of metabolic as opposed to genetic-associated disease, notably including ”lean NAFLD” has recently been recognized. Moreover, NAFLD is a systemic condition, featuring metabolic, cardiovascular and (hepatic/extra-hepatic) cancer risk. Among the clinico-laboratory features of NAFLD we discuss hyperuricemia, insulin resistance, atherosclerosis, gallstones, psoriasis and selected endocrine derangements. NAFLD is a precursor of type 2 diabetes (T2D) and metabolic syndrome and progressive liver disease develops in T2D patients in whom the course of disease is worsened by NAFLD. Finally, lifestyle changes and drug treatment options to be implemented in the individual patient are also critically discussed. In conclusion, this review emphasizes the new concepts on clinical and pathogenic heterogeneity of NAFLD, a systemic disorder with a multifactorial pathogenesis and protean clinical manifestations. It is highly prevalent in certain cohorts of individuals who are thus potentially amenable to selective screening strategies, intensive follow-up schedules for early identification of liver-related and extrahepatic complications and in whom earlier and more aggressive treatment schedules should be carried out whenever possible.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Azienda Ospedaliero-Universitaria di Modena, Ospedale Civile di Baggiovara, 41126 Modena, Italy
| | - Fabio Nascimbeni
- Azienda Ospedaliero-Universitaria di Modena, Ospedale Civile di Baggiovara, 41126 Modena, Italy
- University of Modena and Reggio Emilia, 41126 Modena, Italy
| | - Mauro Maurantonio
- Azienda Ospedaliero-Universitaria di Modena, Ospedale Civile di Baggiovara, 41126 Modena, Italy
| | - Alessandra Marrazzo
- Azienda Ospedaliero-Universitaria di Modena, Ospedale Civile di Baggiovara, 41126 Modena, Italy
- University of Modena and Reggio Emilia, 41126 Modena, Italy
| | - Luca Rinaldi
- Department of Medical, Surgical, Neurological, Geriatric, and Metabolic Sciences, University of Campania “Luigi Vanvitelli”, 80100 Naples, Italy
| | - Luigi Elio Adinolfi
- Department of Medical, Surgical, Neurological, Geriatric, and Metabolic Sciences, University of Campania “Luigi Vanvitelli”, 80100 Naples, Italy
| |
Collapse
|
46
|
Phytosterol esters attenuate hepatic steatosis in rats with non-alcoholic fatty liver disease rats fed a high-fat diet. Sci Rep 2017; 7:41604. [PMID: 28169366 PMCID: PMC5294417 DOI: 10.1038/srep41604] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 12/23/2016] [Indexed: 12/25/2022] Open
Abstract
Given the adverse effects of drugs used for NAFLD treatment, identifying novel and effective natural compound to prevent NAFLD is urgently needed. In the present study, the effects of phytosterol esters (PSEs) on NAFLD were explored. Adult SD rats were randomized into five groups: normal chow diet (NC), high-fat diet (HF), low-, medium- and high-dose PSE treatment plus high-fat diet groups (PSEL, PSEM, and PSEH). Our results showed that the levels of LDL-C in the PSEL group and hepatic TG, TC, and FFA in the three PSEs groups were significantly decreased. Notably, the uric acid (UA) level was significantly decreased by PSEs intervention. The hepatic inflammatory stress was ameliorated via the inhibition of the cytokines, including TGF-β, IL-6, IL-10 and CRP in the PSEs intervention groups. Further, the oxidative status was improved by PSE treatment through adjusting the enzyme activity (SOD and XOD) and decreasing the MDA level. These beneficial effects of PSE may have been partly due to its regulation on the expression of TGF-β1, TGF-β2, TNF-α, UCP-2, PPAR-α and PPAR-γ in hepatic tissue at both mRNA and protein level. The results of this study suggest that PSEs may be used as therapeutic agents for the prevention and progression of NAFLD and that hyperuricemia is induced by high-fat diet consumption.
Collapse
|
47
|
Effect of miglitol on the suppression of nonalcoholic steatohepatitis development and improvement of the gut environment in a rodent model. J Gastroenterol 2017; 52:1180-1191. [PMID: 28349245 PMCID: PMC5666045 DOI: 10.1007/s00535-017-1331-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 03/16/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND The gut environment has been considered to play a role in the development of nonalcoholic steatohepatitis (NASH). α-glucosidase inhibitors (α-GIs) delay carbohydrate absorption and may change the gut environment. We considered that the protective effect of α-GIs against NASH development is related to changes in the gut environment and thus investigated the effects of miglitol, an α-GI, on NASH development and the gut environment. METHODS Mice were divided into three groups and fed a normal chow diet (NCD), a high-fat high-sucrose diet (HFHSD), or HFHSD plus 0.04% miglitol (HFHSD plus M) for 12 weeks. RESULTS Insulin resistance developed more in the HFHSD group than in the NCD group, whereas it was suppressed in the HFHSD plus M group. NASH was evaluated histologically, biochemically, and on the basis of messenger RNA expression levels. Miglitol treatment suppressed HFHSD-induced NASH development with the suppression of hepatic Toll-like receptor 4 expression, increased glucagon-like peptide 1 (GLP-1) concentration, and reduced lipopolysaccharide concentration in portal plasma. Regarding the gut environment, the intestinal transit time was shortened and colon inflammation was suppressed in the HFHSD plus M group compared with the HFHSD group. Regarding the gut microbiota, the abundances of Erysipelotrichaceae and Coriobacteriaceae were increased in the HFHSD group compared with the NCD group, whereas the increase was suppressed in the HFHSD plus M group. CONCLUSIONS We demonstrated that miglitol has a protective effect against HFHSD-induced NASH development. The increased GLP-1 secretion and the suppression of endotoxemia, associated with the changes in the gut environment, including the gut microbiota, could contribute to the underlying mechanisms.
Collapse
|
48
|
Role of Uric Acid Metabolism-Related Inflammation in the Pathogenesis of Metabolic Syndrome Components Such as Atherosclerosis and Nonalcoholic Steatohepatitis. Mediators Inflamm 2016; 2016:8603164. [PMID: 28070145 PMCID: PMC5192336 DOI: 10.1155/2016/8603164] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 11/03/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
Uric acid (UA) is the end product of purine metabolism and can reportedly act as an antioxidant. However, recently, numerous clinical and basic research approaches have revealed close associations of hyperuricemia with several disorders, particularly those comprising the metabolic syndrome. In this review, we first outline the two molecular mechanisms underlying inflammation occurrence in relation to UA metabolism; one is inflammasome activation by UA crystallization and the other involves superoxide free radicals generated by xanthine oxidase (XO). Importantly, recent studies have demonstrated the therapeutic or preventive effects of XO inhibitors against atherosclerosis and nonalcoholic steatohepatitis, which were not previously considered to be related, at least not directly, to hyperuricemia. Such beneficial effects of XO inhibitors have been reported for other organs including the kidneys and the heart. Thus, a major portion of this review focuses on the relationships between UA metabolism and the development of atherosclerosis, nonalcoholic steatohepatitis, and related disorders. Although further studies are necessary, XO inhibitors are a potentially novel strategy for reducing the risk of many forms of organ failure characteristic of the metabolic syndrome.
Collapse
|
49
|
Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: An update. Metabolism 2016; 65:1109-23. [PMID: 27237577 DOI: 10.1016/j.metabol.2016.05.003] [Citation(s) in RCA: 398] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 11/21/2022]
Abstract
Non-alcoholic fatty liver (NAFLD) is the most common liver disease worldwide, progressing from simple steatosis to necroinflammation and fibrosis (leading to non-alcoholic steatohepatitis, NASH), and in some cases to cirrhosis and hepatocellular carcinoma. Inflammation, oxidative stress and insulin resistance are involved in NAFLD development and progression. NAFLD has been associated with several cardiovascular (CV) risk factors including obesity, dyslipidemia, hyperglycemia, hypertension and smoking. NAFLD is also characterized by atherogenic dyslipidemia, postprandial lipemia and high-density lipoprotein (HDL) dysfunction. Most importantly, NAFLD patients have an increased risk for both liver and CV disease (CVD) morbidity and mortality. In this narrative review, the associations between NAFLD, dyslipidemia and vascular disease in NAFLD patients are discussed. NAFLD treatment is also reviewed with a focus on lipid-lowering drugs. Finally, future perspectives in terms of both NAFLD diagnostic biomarkers and therapeutic targets are considered.
Collapse
Affiliation(s)
- Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry (Vascular Disease Prevention Clinics), Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, UK.
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
50
|
Tong W, Ju L, Qiu M, Xie Q, Chen Y, Shen W, Sun W, Wang W, Tian J. Liraglutide ameliorates non-alcoholic fatty liver disease by enhancing mitochondrial architecture and promoting autophagy through the SIRT1/SIRT3-FOXO3a pathway. Hepatol Res 2016; 46:933-43. [PMID: 26666995 DOI: 10.1111/hepr.12634] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/17/2015] [Accepted: 12/05/2015] [Indexed: 12/14/2022]
Abstract
AIM Overwhelming oxidative stress is implicated as crucial in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Liraglutide, a well-established antidiabetes drug, was recently reported to ameliorate NAFLD with an elusive mechanism. We used a mouse model to examine whether liraglutide could ameliorate NAFLD and explored the possible mechanisms. METHODS Twenty C57BL/6J mice were randomly treated with a normal-fat diet or high-fat diet for 16 weeks, then further distributed into four groups and subjected to s.c. injection of liraglutide or saline for 4 weeks. The growth/metabolism, oxidative stress, mitochondrial architecture and autophagy were assessed prospectively at the 20th week. RESULTS High-fat diet inducement resulted in severe NAFLD while liraglutide treatment significantly reversed the trend, marked by reduced bodyweight, improved glucose tolerance and liver triglyceride composition. Reduced hepatic malondialdehyde level, increased mRNA and protein levels of CATALASE and MNSOD indicated liraglutide affected both the oxidative and antioxidative process to ameliorate oxidative stress. After liraglutide administration, the upregulated mRNA and protein levels of mitochondrial fission and fusion-related DRP1, OPA1 and respiratory chain-related COMPLEX1, UCP2 demonstrated the enhancement of mitochondrial architecture which may attenuate the generation of reactive oxygen species (ROS), while the diminished mRNA and protein level of P62 and increased levels of Beclin1 and LC3II/I ratio indicated the promoting autophagy, which probably contribute to the ROS elimination. Further, restored protein levels of Sirtuin1/Sirtuin3 and the downstream p-FOXO3a reveal the probable pathways of liraglutide acting on autophagy. CONCLUSION Liraglutide diminishes oxidative stress by enhancing mitochondrial architecture and promoting autophagy through the SIRT1/SIRT3-FOXO3a-LC3 pathway to ameliorate diet-induced NAFLD.
Collapse
Affiliation(s)
- Wenxin Tong
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Ju
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miaoyan Qiu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qihai Xie
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Chen
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weili Shen
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weihong Sun
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyan Tian
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|