1
|
Abdelrahman RS, Abdelmageed ME. Hepatoprotective effects of the xanthine oxidase inhibitor Febuxostat against thioacetamide-induced liver injury in rats: The role of the Nrf2/ HO-1 and TLR4/ NF-κB pathways. Food Chem Toxicol 2024; 194:115087. [PMID: 39489394 DOI: 10.1016/j.fct.2024.115087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/27/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Experimental models of liver injury have been established utilizing thioacetamide (TAA), a classic liver toxic chemical that causes organ damage via oxidative stress and inflammatory induction. This study examined the impact of Febuxostat (a xanthine oxidase inhibitor; Febu, 10-15 mg/kg, orally) against TAA (500 mg/kg, i.p.) -induced liver injury in rats. Febu significantly attenuated TAA-induced alterations in liver function parameters, in addition to promoting hepatic antioxidant effects through a significant elevation of Heme-oxygenase-1(HO-1), nuclear factor erythroid 2-related factor2 (Nrf2), reduced glutathione (GSH) and superoxide dismutase (SOD) levels and reduction in hepatic malondialdehyde (MDA) content. Moreover, Febu improved the hepatic anti-inflammatory status by increasing the anti-inflammatory cytokine Interleukin (IL-10) level and reducing the levels of the pro-inflammatory cytokines (Nuclear factor kappa B (NF-κB), IL-1β, high-mobility group box1 (HMGB1), receptor for advanced glycation end products (RAGE), and toll-like receptor4 (TLR4) levels, in addition to suppressing the increased protein and mRNA expression levels of tumor necrosis factor alpha (TNF-α) and IL-6, hepatic expression of TNF-α and activated mitogen-activated protein kinases (p-JNK/p-p38 MAPK). Histopathologically, Febu markedly normalized TAA-induced alteration in liver sections. In conclusion, Febu, in a dose-dependent fashion, refines TAA-induced hepatotoxicity by enhancing antioxidant capabilities and decreasing inflammatory signals.
Collapse
Affiliation(s)
- Rehab S Abdelrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taibah University, Al-Madina Al-Munawwarah, 30001, Saudi Arabia
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| |
Collapse
|
2
|
Kraev KI, Geneva-Popova MG, Hristov BK, Uchikov PA, Popova-Belova SD, Kraeva MI, Basheva-Kraeva YM, Stoyanova NS, Mitkova-Hristova VT. Celebrating Versatility: Febuxostat's Multifaceted Therapeutic Application. Life (Basel) 2023; 13:2199. [PMID: 38004339 PMCID: PMC10672185 DOI: 10.3390/life13112199] [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: 10/29/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Febuxostat, initially developed as a xanthine oxidase inhibitor to address hyperuricemia in gout patients, has evolved into a versatile therapeutic agent with multifaceted applications. This review provides a comprehensive overview of febuxostat's mechanism of action, its effectiveness in gout management, its cardiovascular safety profile, renal and hepatic effects, musculoskeletal applications, safety considerations, and emerging research prospects. Febuxostat's primary mechanism involves selective inhibition of xanthine oxidase, resulting in reduced uric acid production. Its pharmacokinetics require personalized dosing strategies based on individual characteristics. In gout management, febuxostat offers a compelling alternative, effectively lowering uric acid levels, relieving symptoms, and supporting long-term control, especially for patients intolerant to allopurinol. Recent studies have demonstrated its cardiovascular safety, and it exhibits minimal hepatotoxicity, making it suitable for those with liver comorbidities. Febuxostat's potential nephroprotective effects and kidney stone prevention properties are noteworthy, particularly for gout patients with renal concerns. Beyond gout, its anti-inflammatory properties hint at applications in musculoskeletal conditions and a broader spectrum of clinical contexts, including metabolic syndrome. Emerging research explores febuxostat's roles in cardiovascular health, neurological disorders, rheumatoid arthritis, and cancer therapy, driven by its anti-inflammatory and antioxidative properties. Future directions include personalized medicine, combination therapies, mechanistic insights, and ongoing long-term safety monitoring, collectively illuminating the promising landscape of febuxostat's multifaceted therapeutic potential.
Collapse
Affiliation(s)
- Krasimir Iliev Kraev
- Department of Propedeutics of Internal Diseases, Medical Faculty, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| | | | - Bozhidar Krasimirov Hristov
- Second Department of Internal Diseases, Medical Faculty, Medical University of Plovdiv, 6000 Plovdiv, Bulgaria
| | - Petar Angelov Uchikov
- Department of Special Surgery, Medical Faculty, Medical University of Plovdiv, 6000 Plovdiv, Bulgaria
| | | | - Maria Ilieva Kraeva
- Department of Otorhynolaryngology, Medical Faculty, Medical University of Plovdiv, 6000 Plovdiv, Bulgaria
| | - Yordanka Mincheva Basheva-Kraeva
- Department of Ophthalmology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
- University Eye Clinic, University Hospital, 4000 Plovdiv, Bulgaria
| | - Nina Staneva Stoyanova
- Department of Ophthalmology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
- University Eye Clinic, University Hospital, 4000 Plovdiv, Bulgaria
| | - Vesela Todorova Mitkova-Hristova
- Department of Ophthalmology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
- University Eye Clinic, University Hospital, 4000 Plovdiv, Bulgaria
| |
Collapse
|
3
|
Feaver RE, Bowers MS, Cole BK, Hoang S, Lawson MJ, Taylor J, LaMoreaux BD, Zhao L, Henke BR, Johns BA, Nyborg AC, Wamhoff BR, Figler RA. Human cardiovascular disease model predicts xanthine oxidase inhibitor cardiovascular risk. PLoS One 2023; 18:e0291330. [PMID: 37682977 PMCID: PMC10490929 DOI: 10.1371/journal.pone.0291330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Some health concerns are often not identified until late into clinical development of drugs, which can place participants and patients at significant risk. For example, the United States Food and Drug Administration (FDA) labeled the xanthine oxidase inhibitor febuxostat with a"boxed" warning regarding an increased risk of cardiovascular death, and this safety risk was only identified during Phase 3b clinical trials after its approval. Thus, better preclinical assessment of drug efficacy and safety are needed to accurately evaluate candidate drug risk earlier in discovery and development. This study explored whether an in vitro vascular model incorporating human vascular cells and hemodynamics could be used to differentiate the potential cardiovascular risk associated with molecules that have similar on-target mechanisms of action. We compared the transcriptomic responses induced by febuxostat and other xanthine oxidase inhibitors to a database of 111 different compounds profiled in the human vascular model. Of the 111 compounds in the database, 107 are clinical-stage and 33 are FDA-labelled for increased cardiovascular risk. Febuxostat induces pathway-level regulation that has high similarity to the set of drugs FDA-labelled for increased cardiovascular risk. These results were replicated with a febuxostat analog, but not another structurally distinct xanthine oxidase inhibitor that does not confer cardiovascular risk. Together, these data suggest that the FDA warning for febuxostat stems from the chemical structure of the medication itself, rather than the target, xanthine oxidase. Importantly, these data indicate that cardiovascular risk can be evaluated in this in vitro human vascular model, which may facilitate understanding the drug candidate safety profile earlier in discovery and development.
Collapse
Affiliation(s)
- Ryan E. Feaver
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - M. Scott Bowers
- Horizon Therapeutics plc, Deerfield, Illinois, United States of America
| | - Banumathi K. Cole
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Steve Hoang
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Mark J. Lawson
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Justin Taylor
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | | | - Lin Zhao
- Horizon Therapeutics plc, Deerfield, Illinois, United States of America
| | - Brad R. Henke
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Brian A. Johns
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Andrew C. Nyborg
- Horizon Therapeutics plc, Deerfield, Illinois, United States of America
| | - Brian R. Wamhoff
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| | - Robert A. Figler
- HemoShear Therapeutics, Incorporated., Charlottesville, Virginia, United States of America
| |
Collapse
|
4
|
Burrage EN, Coblentz T, Prabhu SS, Childers R, Bryner RW, Lewis SE, DeVallance E, Kelley EE, Chantler PD. Xanthine oxidase mediates chronic stress-induced cerebrovascular dysfunction and cognitive impairment. J Cereb Blood Flow Metab 2023; 43:905-920. [PMID: 36655326 PMCID: PMC10196752 DOI: 10.1177/0271678x231152551] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/20/2023]
Abstract
Xanthine oxidase (XO) mediates vascular function. Chronic stress impairs cerebrovascular function and increases the risk of stroke and cognitive decline. Our study determined the role of XO on stress-induced cerebrovascular dysfunction and cognitive decline. We measured middle cerebral artery (MCA) function, free radical formation, and working memory in 6-month-old C57BL/6 mice who underwent 8 weeks of control conditions or unpredictable chronic mild stress (UCMS) with or without febuxostat (50 mg/L), a XO inhibitor. UCMS mice had an impaired MCA dilation to acetylcholine vs. controls (p < 0.0001), and increased total free radical formation, XOR protein levels, and hydrogen peroxide production in the liver compared to controls. UCMS increased hydrogen peroxide production in the brain and cerebrovasculature compared to controls. Working memory, using the y-maze test, was impaired (p < 0.05) in UCMS mice compared to control mice. However, blocking XO using febuxostat prevented the UCMS-induced impaired MCA response, while free radical production and hydrogen peroxide levels were similar to controls in the liver and brain of UCMS mice treated with febuxostat. Further, UCMS + Feb mice did not have a significant reduction in working memory. These data suggest that the cerebrovascular dysfunction associated with chronic stress may be driven by XO, which leads to a reduction in working memory.
Collapse
Affiliation(s)
- Emily N Burrage
- Department of Neuroscience, West
Virginia University School of Medicine, Morgantown, WV, USA
| | - Tyler Coblentz
- Division of Exercise Physiology,
West Virginia University School of Medicine, Morgantown, WV, USA
| | - Saina S Prabhu
- Department of Pharmaceutical
Sciences, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Ryan Childers
- Division of Exercise Physiology,
West Virginia University School of Medicine, Morgantown, WV, USA
| | - Randy W Bryner
- Division of Exercise Physiology,
West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sarah E Lewis
- Department of Physiology and
Pharmacology, West Virginia University School of Medicine, Morgantown, WV,
USA
| | - Evan DeVallance
- Department of Physiology and
Pharmacology, West Virginia University School of Medicine, Morgantown, WV,
USA
| | - Eric E Kelley
- Department of Physiology and
Pharmacology, West Virginia University School of Medicine, Morgantown, WV,
USA
| | - Paul D Chantler
- Department of Neuroscience, West
Virginia University School of Medicine, Morgantown, WV, USA
- Division of Exercise Physiology,
West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
5
|
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
|
6
|
Wang Q, Qi H, Wu Y, Yu L, Bouchareb R, Li S, Lassén E, Casalena G, Stadler K, Ebefors K, Yi Z, Shi S, Salem F, Gordon R, Lu L, Williams RW, Duffield J, Zhang W, Itan Y, Böttinger E, Daehn I. Genetic susceptibility to diabetic kidney disease is linked to promoter variants of XOR. Nat Metab 2023; 5:607-625. [PMID: 37024752 PMCID: PMC10821741 DOI: 10.1038/s42255-023-00776-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/07/2023] [Indexed: 04/08/2023]
Abstract
The lifetime risk of kidney disease in people with diabetes is 10-30%, implicating genetic predisposition in the cause of diabetic kidney disease (DKD). Here we identify an expression quantitative trait loci (QTLs) in the cis-acting regulatory region of the xanthine dehydrogenase, or xanthine oxidoreductase (Xor), a binding site for C/EBPβ, to be associated with diabetes-induced podocyte loss in DKD in male mice. We examine mouse inbred strains that are susceptible (DBA/2J) and resistant (C57BL/6J) to DKD, as well as a panel of recombinant inbred BXD mice, to map QTLs. We also uncover promoter XOR orthologue variants in humans associated with high risk of DKD. We introduced the risk variant into the 5'-regulatory region of XOR in DKD-resistant mice, which resulted in increased Xor activity associated with podocyte depletion, albuminuria, oxidative stress and damage restricted to the glomerular endothelium, which increase further with type 1 diabetes, high-fat diet and ageing. Therefore, differential regulation of Xor contributes to phenotypic consequences with diabetes and ageing.
Collapse
Affiliation(s)
- Qin Wang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Haiying Qi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yiming Wu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Liping Yu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rihab Bouchareb
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shuyu Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emelie Lassén
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriella Casalena
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Krisztian Stadler
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Kerstin Ebefors
- Department of Neuroscience and Physiology, Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Zhengzi Yi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shaolin Shi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fadi Salem
- Pathology, Molecular and Cell based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Gordon
- Pathology, Molecular and Cell based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuval Itan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erwin Böttinger
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Hasso Plattner Institute for Digital Heath at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Hasso Plattner Institut, University of Potsdam, Potsdam, Germany
| | - Ilse Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
7
|
Ramírez-Velásquez I, Bedoya-Calle ÁH, Vélez E, Caro-Lopera FJ. Shape Theory Applied to Molecular Docking and Automatic Localization of Ligand Binding Pockets in Large Proteins. ACS OMEGA 2022; 7:45991-46002. [PMID: 36570297 PMCID: PMC9773186 DOI: 10.1021/acsomega.2c02227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 10/11/2022] [Indexed: 06/17/2023]
Abstract
Automatic search of cavities and binding mode analysis between a ligand and a 3D protein receptor are challenging problems in drug design or repositioning. We propose a solution based on a shape theory theorem for an invariant coupled system of ligand-protein. The theorem provides a matrix representation with the exact formulas to be implemented in an algorithm. The method involves the following results: (1) exact formulae for the shape coordinates of a located-rotated invariant coupled system; (2) a parameterized search based on a suitable domain of van der Waals radii; (3) a scoring function for the discrimination of sites by measuring the distance between two invariant coupled systems including the atomic mass; (4) a matrix representation of the Lennard-Jones potential type 6-12 and 6-10 as the punctuation function of the algorithm for a molecular docking; and (5) the optimal molecular docking as a solution of an optimization problem based on the exploration of an exhaustive set of rotations. We apply the method in the xanthine oxidase protein with the following ligands: hypoxanthine, febuxostat, and chlorogenic acid. The results show automatic cavity detection and molecular docking not assisted by experts with meaningful amino acid interactions. The method finds better affinities than the expert software for known published cavities.
Collapse
Affiliation(s)
- Iliana Ramírez-Velásquez
- Faculty
of Exact and Applied Sciences, Instituto
Tecnológico Metropolitano ITM, Cll. 73 # 76A-354, Medellín050034, Colombia
- Doctorate
in Modeling and Scientific Computing, Faculty of Basic Sciences, University of Medellin, Medellin050026, Colombia
| | - Álvaro H. Bedoya-Calle
- Faculty
of Basic Sciences, University of Medellin, Cra. 87 # 30-65, Medellín050026, Colombia
| | - Ederley Vélez
- Faculty
of Basic Sciences, University of Medellin, Cra. 87 # 30-65, Medellín050026, Colombia
| | | |
Collapse
|
8
|
Quy PT, Dzung NA, Van Bay M, Van Bon N, Dung DM, Nam PC, Thong NM. Insights into antiradical mechanism and pro-oxidant enzyme inhibitor activity of walterolactone A/B 6- O-gallate-β-d-pyranoglucoside originating from Euonymus laxiflorus Champ. using in silico study. RSC Adv 2022; 12:29975-29982. [PMID: 36321076 PMCID: PMC9580510 DOI: 10.1039/d2ra05312h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
The ability of a new compound, Wal, (walterolactone A/B 6-O-gallate-β-d-pyranoglucoside) originating from Euonymus laxiflorus Champ. as a hydroperoxyl radical scavenger and pro-oxidant enzyme inhibitor was studied in silico. Different mechanisms, reaction locations, and chemical species of Wal in aqueous solution were taken into consideration. Formal hydrogen transfer from the OH group has been discovered as the chemical process that contributes most to the antioxidant properties of Wal in nonpolar and aqueous solutions. The overall rate coefficients for polar and non-polar environments are expected to have values of 7.85 × 106 M-1 s-1 and 4.84 × 105 M-1 s-1, respectively. According to the results of the investigation, Wal has greater scavenging activity against the HOO˙ radical than the reference antioxidant Trolox at physiological pH (7.4). In addition, docking results indicate that Wal's antioxidant properties involve the inhibition of the activity of enzyme families (CP450, MP, NO, and XO) that are responsible for ROS production.
Collapse
Affiliation(s)
- Phan Tu Quy
- Department of Natural Sciences & Technology, Tay Nguyen University Buon Ma Thuot 630000 Vietnam
| | - Nguyen Anh Dzung
- Institute of Biotechnology and Environment, Tay Nguyen University Buon Ma Thuot 630000 Vietnam
| | - Mai Van Bay
- The University of Danang - University of Science and Education Danang 550000 Vietnam
| | - Nguyen Van Bon
- Institute of Biotechnology and Environment, Tay Nguyen University Buon Ma Thuot 630000 Vietnam
| | - Doan Manh Dung
- Institute of Biotechnology and Environment, Tay Nguyen University Buon Ma Thuot 630000 Vietnam
| | - Pham Cam Nam
- The University of Danang - University of Science and Technology Danang 550000 Vietnam
| | - Nguyen Minh Thong
- The University of Danang - Campus in Kon Tum 704 Phan Dinh Phung Kon Tum Vietnam
| |
Collapse
|
9
|
Skeletal muscle as a reservoir for nitrate and nitrite: The role of xanthine oxidase reductase (XOR). Nitric Oxide 2022; 129:102-109. [DOI: 10.1016/j.niox.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/16/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
|
10
|
Cam Nam P, Van Bay M, Vo QV, Mechler A, Minh Thong N. Tautomerism and antioxidant power of sulfur-benzo[h]quinoline: DFT and molecular docking studies. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
11
|
Alqahtani A, Chidambara K, Asseri K, Venkatesan K, Ahmed Qure A, Aldahish A, Alqahtani T, Alghazwani Y, Bin Emran T, Hassan HM, Asiri SA, Abdulla Kh N, Balakumar P. Renoprotective and in silico Modeling Studies of Febuxostat in Gentamicin Induced Nephrotoxic Rats. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.994.1003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
12
|
Butts B, Brown JA, Denney TS, Ballinger S, Lloyd SG, Oparil S, Sanders P, Merriman TR, Gaffo A, Singh J, Kelley EE, Calhoun DA, Dell'Italia LJ. Racial Differences in XO (Xanthine Oxidase) and Mitochondrial DNA Damage-Associated Molecular Patterns in Resistant Hypertension. Hypertension 2022; 79:775-784. [PMID: 35164526 PMCID: PMC10652275 DOI: 10.1161/hypertensionaha.121.18298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/22/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND We previously reported increased plasma XO (xanthine oxidase) activity in patients with resistant hypertension. Increased XO can cause mitochondrial DNA damage and promote release of fragments called mitochondrial DNA damage-associated molecular patterns (mtDNA DAMPs). Here, we report racial differences in XO activity and mtDNA DAMPs in Black and White adults with resistant hypertension. METHODS This retrospective study includes 91 resistant hypertension patients (44% Black, 47% female) with blood pressure >140/90 mm Hg on ≥4 medications and 37 normotensive controls (30% Black, 54% female) with plasma XO activity, mtDNA DAMPs, and magnetic resonance imaging of left ventricular morphology and function. RESULTS Black-resistant hypertension patients were younger (mean age 52±10 versus 59±10 years; P=0.001), with higher XO activity and left ventricular wall thickness, and worse diastolic dysfunction than White resistant hypertension patients. Urinary sodium excretion (mg/24 hour per kg) was positively related to left ventricular end-diastolic volume (r=0.527, P=0.001) and left ventricular mass (r=0.394, P=0.02) among Black but not White resistant hypertension patients. Patients with resistant hypertension had increased mtDNA DAMPs versus controls (P<0.001), with Black mtDNA DAMPS greater than Whites (P<0.001). Transmission electron microscopy of skeletal muscle biopsies in resistant hypertension patients demonstrates mitochondria cristae lysis, myofibrillar loss, large lipid droplets, and glycogen accumulation. CONCLUSIONS These data warrant a large study to examine the role of XO and mitochondrial mtDNA DAMPs in cardiac remodeling and heart failure in Black adults with resistant hypertension.
Collapse
Affiliation(s)
- Brittany Butts
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
| | - Jamelle A Brown
- Center for Free Radical Biology and Department of Pathology, UAB SOM (J.A.B., S.B.)
| | - Thomas S Denney
- Department of Electrical and Computer Engineering, Auburn University (T.S.D.)
| | - Scott Ballinger
- Center for Free Radical Biology and Department of Pathology, UAB SOM (J.A.B., S.B.)
| | - Steven G Lloyd
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| | - Suzanne Oparil
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
| | - Paul Sanders
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
- Nephrology Research and Training Center and Division of Nephrology UAB SOM (P.S.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, UAB SOM (T.R.M., A.G., J.S.)
| | - Angelo Gaffo
- Division of Clinical Immunology and Rheumatology, UAB SOM (T.R.M., A.G., J.S.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| | - Jasvinder Singh
- Division of Clinical Immunology and Rheumatology, UAB SOM (T.R.M., A.G., J.S.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University (E.E.K.)
| | - David A Calhoun
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
| | - Louis J Dell'Italia
- Division of Cardiovascular Disease, University of Alabama at Birmingham (UAB) School of Medicine (SOM) (B.B., S.G.L., S.O., P.S., D.A.C., L.J.D.)
- Birmingham Department of Veterans Affairs Health Care System (S.G.L., P.S., A.G., J.S., L.J.D.)
| |
Collapse
|
13
|
Mechanism of Antioxidant Activity of Betanin, Betanidin and Respective C15-Epimers via Shape Theory, Molecular Dynamics, Density Functional Theory and Infrared Spectroscopy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27062003. [PMID: 35335368 PMCID: PMC8954076 DOI: 10.3390/molecules27062003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022]
Abstract
Betanin and betanidin are compounds with extensive interest; they are effectively free radical scavengers. The present work aims to elucidate the differences between the mechanism of the antioxidant activity of betanin, betanidin, and their respective C15-epimers. Shape Theory establishes comparisons between the molecules’ geometries and determines parallelisms with the descriptors BDE, PA, ETE IP, PDE, and infrared spectra (IR) obtained from the molecule simulations. Furthermore, the molecules were optimized using the B3LYP/6-31+G(d,p) protocol. Finally, the molecular docking technique analyzes the antioxidant activity of the compounds in the complex with the therapeutic target xanthine oxidase (XO), based on a new proposal for the geometrical arrangement of the ligand atoms in the framework of Shape Theory. The results obtained indicate that the SPLET mechanism is the most favorable in all the molecules studied and that the first group that loses the hydrogen atom in the four molecules is the C17COOH, presenting less PA the isobetanidin. Furthermore, regarding the molecular docking, the interactions of these compounds with the target were favorable, standing out to a greater extent the interactions of isobetanidin with XO, which were analyzed after applying molecular dynamics.
Collapse
|
14
|
Topiroxostat versus allopurinol in patients with chronic heart failure complicated by hyperuricemia: A prospective, randomized, open-label, blinded-end-point clinical trial. PLoS One 2022; 17:e0261445. [PMID: 35077456 PMCID: PMC8789120 DOI: 10.1371/journal.pone.0261445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background The benefits of xanthine oxidase inhibitors to chronic heart failure (CHF) patients is controversial. We investigated the beneficial effects of a novel xanthine oxidoreductase inhibitor, topiroxostat, in patients with CHF and hyperuricemia (HU), in comparison to allopurinol. Methods and results The prospective, randomized open-label, blinded-end-point study was performed in 141 patients with CHF and HU at 4 centers. Patients were randomly assigned to either topiroxostat or allopurinol group to achieve target uric acid level ≤6.0 mg/dL. According to the protocol, 140 patients were followed up for 24 weeks. Percent change in ln (N-terminal-proB-type natriuretic peptide) at week 24 (primary endpoint) was comparable between topiroxostat and allopurinol groups (1.6±8.2 versus -0.4±8.0%; P = 0.17). In the limited number of patients with heart failure with reduced ejection fraction (HFrEF) (left ventricle ejection fraction <45%), ratio of peak early diastolic flow velocity at mitral valve leaflet to early diastolic mitral annular motion velocity (E/e’) decreased in topiroxostat group, but not in allopurinol group. Urinary 8-hydroxy-2’-deoxyguanosine and L-type fatty acid-binding protein levels increased and osmolality decreased significantly in allopurinol group, while these changes were less or absent in topiroxostat group. In allopurinol group HFrEF patients, additional to the increases in these urinary marker levels, urinary creatinine levels decreased, with no change in clearance, but not in topiroxostat group. Conclusions Compared with allopurinol, topiroxostat did not show great benefits in patients with CHF and HU. However, topiroxostat might have potential advantages of reducing left ventricular end-diastolic pressure, not worsening oxidative stress in proximal renal tubule, and renoprotection over allopurinol in HFrEF patients.
Collapse
|
15
|
Suzuki S, Yoshihisa A, Yokokawa T, Kobayashi A, Yamaki T, Kunii H, Nakazato K, Tsuda A, Tsuda T, Ishibashi T, Konno I, Yamaguchi O, Machii H, Nozaki N, Niizeki T, Miyamoto T, Takeishi Y. Comparison between febuxostat and allopurinol uric acid-lowering therapy in patients with chronic heart failure and hyperuricemia: a multicenter randomized controlled trial. J Int Med Res 2021; 49:3000605211062770. [PMID: 34914568 PMCID: PMC8689623 DOI: 10.1177/03000605211062770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objective Heart failure (HF) is a common and highly morbid cardiovascular disorder. Oxidative stress worsens HF, and uric acid (UA) is a useful oxidative stress marker. The novel anti-hyperuricemic drug febuxostat is a potent non-purine selective xanthine oxidase inhibitor. The present study examined the UA-lowering and prognostic effects of febuxostat in patients with HF compared with conventional allopurinol. Methods This multicenter, randomized trial included 263 patients with chronic HF who were randomly assigned to two groups and received allopurinol or febuxostat (UA >7.0 mg/dL). All patients were followed up for 3 years after enrollment. Results There were no significant differences in baseline clinical characteristics between the two groups. The UA level was significantly decreased after 3 years of drug administration compared with the baseline in both groups. Urine levels of the oxidative stress marker 8-hydroxy-2′-deoxyguanosine were lower in the febuxostat group than in the allopurinol group (11.0 ± 9.6 vs. 22.9 ± 15.9 ng/mL), and the rate of patients free from hospitalization due to worsening HF tended to be higher in the febuxostat group than in the allopurinol group (89.0% vs. 83.0%). Conclusions Febuxostat is potentially more effective than allopurinol for treating patients with chronic HF and hyperuricemia. This study was registered in the University Hospital Medical Information Network Clinical Trials Registry (https://www.umin.ac.jp/ctr/; ID: 000009817).
Collapse
Affiliation(s)
- Satoshi Suzuki
- Department of Cardiovascular Medicine, 12775Fukushima Medical University, Fukushima Medical University, Fukushima, Japan.,Cardiology Department, 13881Takeda General Hospital, Takeda General Hospital, Aizuwakamatsu, Japan
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, 12775Fukushima Medical University, Fukushima Medical University, Fukushima, Japan
| | - Tetsuro Yokokawa
- Department of Cardiovascular Medicine, 12775Fukushima Medical University, Fukushima Medical University, Fukushima, Japan
| | - Atsushi Kobayashi
- Department of Cardiovascular Medicine, 12775Fukushima Medical University, Fukushima Medical University, Fukushima, Japan
| | - Takayoshi Yamaki
- Department of Cardiovascular Medicine, 12775Fukushima Medical University, Fukushima Medical University, Fukushima, Japan
| | - Hiroyuki Kunii
- Department of Cardiovascular Medicine, 12775Fukushima Medical University, Fukushima Medical University, Fukushima, Japan
| | - Kazuhiko Nakazato
- Department of Cardiovascular Medicine, 12775Fukushima Medical University, Fukushima Medical University, Fukushima, Japan
| | - Akihiro Tsuda
- Cardiology Department, 274894Sukagawa Hospital, 274894Sukagawa Hospital, Sukagawa, Japan
| | - Tatsunori Tsuda
- Cardiology Department, 274894Sukagawa Hospital, 274894Sukagawa Hospital, Sukagawa, Japan
| | - Toshiyuki Ishibashi
- Department of Cardiovascular Medicine, 36952Ohara General Hospital, Ohara General Hospital, Fukushima, Japan
| | - Ichiro Konno
- Department of Cardiovascular Medicine, 36952Ohara General Hospital, Ohara General Hospital, Fukushima, Japan
| | - Osamu Yamaguchi
- Department of Cardiovascular Medicine, 36952Ohara General Hospital, Ohara General Hospital, Fukushima, Japan
| | - Hirofumi Machii
- Department of Cardiovascular Medicine, 36952Ohara General Hospital, Ohara General Hospital, Fukushima, Japan
| | - Naoki Nozaki
- Cardiology Department, Ayase Heart Hospital, Tokyo, Japan
| | - Takeshi Niizeki
- Department of Cardiology, 50191Okitama Public General Hospital, 50191Okitama Public General Hospital, Kawanishi, Japan
| | - Takuya Miyamoto
- First Department of Internal Medicine, 538443Yamagata University Hospital, Yamagata University Hospital, Yamagata, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, 12775Fukushima Medical University, Fukushima Medical University, Fukushima, Japan
| | | |
Collapse
|
16
|
Odake K, Tsujii M, Iino T, Chiba K, Kataoka T, Sudo A. Febuxostat treatment attenuates oxidative stress and inflammation due to ischemia-reperfusion injury through the necrotic pathway in skin flap of animal model. Free Radic Biol Med 2021; 177:238-246. [PMID: 34737143 DOI: 10.1016/j.freeradbiomed.2021.10.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Ischemia-reperfusion (I/R) injury is a major contributor to skin flap necrosis, which is a serious complication of reconstructive surgery. The purpose of this study was to evaluate the protective effect of treatment with febuxostat, a selective xanthine oxidase inhibitor, on I/R injury in the skin flap of an animal (rat) model. METHODS Superficial epigastric flaps were raised in Sprague-Dawley rats and subjected to ischemia for 3 h. Febuxostat at a dose of 10 mg/kg/day was administered to rats in drinking water from 1 week before the surgery (Feb group). Control animals received no drugs (Con group). The mean ratio of flap survival and contraction was evaluated and compared between animals with and without administration of febuxostat on day 5 after the surgery. In addition, infiltration by polymorphonuclear leukocytes and muscles of the panniculus carnosus in the flap were histologically evaluated using hematoxylin-eosin staining. Furthermore, xanthine oxidase activity, ATP levels, superoxide dismutase activity, and expression of 8-hydroxy-2'-deoxyguanosine (8-OHdG), tumor necrosis factor-α, and interleukin-1β were quantitatively assessed in the skin flap 24 h after the surgery. RESULTS In the Feb group, the survival and contraction rates at the 5 d timepoint post-surgery were significantly higher and lower than those in the Con group, respectively. Histological analysis showed significant reduction in polymorphonuclear leukocyte infiltration and muscle injury scores due to I/R injury in the Feb group. The expression of 8-OHdG was also significantly inhibited in animals administered febuxostat. Biochemical analysis showed a significant reduction in xanthine oxidase activity and significant increases in ATP levels and superoxide dismutase activity in the Feb group. Furthermore, the expression of interleukin-1β was significantly lower in the Feb group than in the Con group. CONCLUSION Febuxostat, which is clinically used for the treatment of hyperuricemia, was effective against necrosis of the skin flap via inhibition of oxidative stress and inflammation caused by I/R injury.
Collapse
Affiliation(s)
- Kazuya Odake
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Japan
| | - Masaya Tsujii
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Japan.
| | - Takahiro Iino
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Japan
| | - Katsura Chiba
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Japan
| | - Takeshi Kataoka
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Graduate School of Medicine, Mie University, Japan
| |
Collapse
|
17
|
Xu D, Murakoshi N, Tajiri K, Duo F, Okabe Y, Murakata Y, Yuan Z, Li S, Aonuma K, Song Z, Shimoda Y, Mori H, Sato A, Nogami A, Aonuma K, Ieda M. Xanthine oxidase inhibitor febuxostat reduces atrial fibrillation susceptibility by inhibition of oxidized CaMKII in Dahl salt-sensitive rats. Clin Sci (Lond) 2021; 135:2409-2422. [PMID: 34386810 DOI: 10.1042/cs20210405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022]
Abstract
Oxidative stress could be a possible mechanism and a therapeutic target of atrial fibrillation (AF). However, the effects of the xanthine oxidase (XO) inhibition for AF remain to be fully elucidated. We investigated the effects of a novel XO inhibitor febuxostat on AF compared with allopurinol in hypertension rat model. Five-week-old Dahl salt-sensitive rats were fed either low-salt (LS) (0.3% NaCl) or high-salt (HS) (8% NaCl) diet. After 4 weeks of diet, HS diet rats were divided into three groups: orally administered to vehicle (HS-C), febuxostat (5 mg/kg/day) (HS-F), or allopurinol (50 mg/kg/day) (HS-A). After 4 weeks of treatment, systolic blood pressure (SBP) was significantly higher in HS-C than LS, and it was slightly but significantly decreased by treatment with each XO inhibitor. AF duration was significantly prolonged in HS-C compared with LS, and significantly suppressed in both HS-F and HS-A (LS; 5.8 ± 3.5 s, HS-C; 33.9 ± 23.7 s, HS-F; 15.0 ± 14.1 s, HS-A; 20.1 ± 11.9 s: P<0.05). Ca2+ spark frequency was obviously increased in HS-C rats and reduced in the XO inhibitor-treated rats, especially in HS-F group. Western blotting revealed that the atrial expression levels of Met281/282-oxidized Ca2+/Calmodulin-dependent kinase II (CaMKII) and Ser2814-phosphorylated ryanodine receptor 2 were significantly increased in HS-C, and those were suppressed in HS-F and HS-A. Decreased expression of gap junction protein connexin 40 in HS-C was partially restored by treatment with each XO inhibitor. In conclusion, XO inhibitor febuxostat, as well as allopurinol, could reduce hypertension-related increase in AF perpetuation by restoring Ca2+ handling and gap junction.
Collapse
Affiliation(s)
- DongZhu Xu
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Feng Duo
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuta Okabe
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiko Murakata
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zixun Yuan
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Siqi Li
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuhiro Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Zonghu Song
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuzuno Shimoda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruka Mori
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Sato
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazutaka Aonuma
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
18
|
Lewis SE, Rosencrance CB, De Vallance E, Giromini A, Williams XM, Oh JY, Schmidt H, Straub AC, Chantler PD, Patel RP, Kelley EE. Human and rodent red blood cells do not demonstrate xanthine oxidase activity or XO-catalyzed nitrite reduction to NO. Free Radic Biol Med 2021; 174:84-88. [PMID: 34273539 PMCID: PMC9257433 DOI: 10.1016/j.freeradbiomed.2021.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/18/2022]
Abstract
A number of molybdopterin enzymes, including xanthine oxidoreductase (XOR), aldehyde oxidase (AO), sulfite oxidase (SO), and mitochondrial amidoxime reducing component (mARC), have been identified as nitrate and nitrite reductases. Of these enzymes, XOR has been the most extensively studied and reported to be a substantive source of nitric oxide (NO) under inflammatory/hypoxic conditions that limit the catalytic activity of the canonical NOS pathway. It has also been postulated that XOR nitrite reductase activity extends to red blood cell (RBCs) membranes where it has been immunohistochemically identified. These findings, when combined with countervailing reports of XOR activity in RBCs, incentivized our current study to critically evaluate XOR protein abundance/enzymatic activity in/on RBCs from human, mouse, and rat sources. Using various protein concentrations of RBC homogenates for both human and rodent samples, neither XOR protein nor enzymatic activity (xanthine → uric acid) was detectable. In addition, potential loading of RBC-associated glycosaminoglycans (GAGs) by exposing RBC preparations to purified XO before washing did not solicit detectable enzymatic activity (xanthine → uric acid) or alter NO generation profiles. To ensure these observations extended to absence of XOR-mediated contributions to overall RBC-associated nitrite reduction, we examined the nitrite reductase activity of washed and lysed RBC preparations via enhanced chemiluminescence in the presence or absence of the XOR-specific inhibitor febuxostat (Uloric®). Neither addition of inhibitor nor the presence of the XOR substrate xanthine significantly altered the rates of nitrite reduction to NO by RBC preparations from either human or rodent sources confirming the absence of XO enzymatic activity. Furthermore, examination of the influence of the age (young cells vs. old cells) of human RBCs on XO activity also failed to demonstrate detectable XO protein. Combined, these data suggest: 1) that XO does not contribute to nitrite reduction in/on human and rodent erythrocytes, 2) care should be taken to validate immuno-detectable XO by demonstrating enzymatic activity, and 3) XO does not associate with human erythrocytic glycosaminoglycans or participate in nonspecific binding.
Collapse
Affiliation(s)
- Sara E Lewis
- West Virginia University Departments of Physiology and Pharmacology, USA
| | | | - Evan De Vallance
- West Virginia University Departments of Physiology and Pharmacology, USA
| | - Andrew Giromini
- West Virginia University Departments of Physiology and Pharmacology, USA
| | - Xena M Williams
- West Virginia University Departments of Physiology and Pharmacology, USA
| | - Joo-Yeun Oh
- University of Alabama at Birmingham Center for Free Radical Biology, USA
| | - Heidi Schmidt
- University of Pittsburgh Vascular Medicine Institute, USA
| | - Adam C Straub
- University of Pittsburgh Vascular Medicine Institute, USA
| | | | - Rakesh P Patel
- University of Alabama at Birmingham Center for Free Radical Biology, USA
| | - Eric E Kelley
- West Virginia University Departments of Physiology and Pharmacology, USA.
| |
Collapse
|
19
|
Tsukamoto T, Tsujii M, Odake K, Iino T, Nakamura T, Matsumine A, Sudo A. Febuxostat reduces muscle wasting in tumor-bearing mice with LM8 osteosarcoma cells via inhibition of reactive oxygen species generation. Free Radic Res 2021; 55:810-820. [PMID: 34278932 DOI: 10.1080/10715762.2021.1947502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cachexic condition due to malignant tumors has been a challenging problem. The aim of this study is to analyze effects of febuxostat on both in vitro and in vivo models of the wasting of skeletal muscles, due to LM8 osteosarcoma cells. C2C12 myotubes were incubated in the conditioned medium of LM8. Febuxostat was added at a concentration of 3 µM and 30 µM, and ROS, diameter of myotubes, and expression of atrogin-1 were analyzed. Furthermore, an in vivo study was performed by subcutaneous injection of LM8 on C3H mice. Febuxostat was administered in the drinking water at 5 µg/ml, and 25 µg/ml. In addition, tumor-bearing mice without febuxostat (group TB) and control mice (group C) were established. At 4 weeks, body weight, wet weights of the gastrocnemius muscles, XO activity, 8-OHdG, and expression of TNF-α and IL-6 were evaluated. ROS generation, atrophy of myotubes, and upregulation of atrogin-1 were clearly observed in C2C12 myotubes following incubation in the conditioned medium. These pathological conditions were significantly inhibited by febuxostat administration. Furthermore, mice in group TB showed significant loss of body weight and muscle weight in which XO activity, 8-OHdG, and expression of IL-6 were significantly increased compared to those in group C. Febuxostat administration not only significantly improved the body weight and muscleweight, but also reduced markers of oxidative stress and pro-inflammatory cytokines. Febuxostat did not show anti-tumor effects. Febuxostat, which is clinically used for treatment of hyperuricemia, is effective against the wasting of the skeletal muscles induced by LM8 osteosarcoma cells.
Collapse
Affiliation(s)
- Tadashi Tsukamoto
- Department of Orthopaedic Surgery, Mie university Graduate School of Medicine, Tsu, Japan
| | - Masaya Tsujii
- Department of Orthopaedic Surgery, Mie university Graduate School of Medicine, Tsu, Japan
| | - Kazuya Odake
- Department of Orthopaedic Surgery, Mie university Graduate School of Medicine, Tsu, Japan
| | - Takahiro Iino
- Department of Orthopaedic Surgery, Mie university Graduate School of Medicine, Tsu, Japan
| | - Tomoki Nakamura
- Department of Orthopaedic Surgery, Mie university Graduate School of Medicine, Tsu, Japan
| | - Akihiko Matsumine
- Department of Orthopaedic Surgery, Fukui University Faculty of Medical Science, Eiheiji-Cho, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Mie university Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
20
|
Martorell M, Lucas X, Alarcón-Zapata P, Capó X, Quetglas-Llabrés MM, Tejada S, Sureda A. Targeting Xanthine Oxidase by Natural Products as a Therapeutic Approach for Mental Disorders. Curr Pharm Des 2021; 27:367-382. [PMID: 32564744 DOI: 10.2174/1381612826666200621165839] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/08/2020] [Indexed: 11/22/2022]
Abstract
Mental disorders comprise diverse human pathologies, including depression, bipolar affective disorder, schizophrenia, and dementia that affect millions of people around the world. The causes of mental disorders are unclear, but growing evidence suggests that oxidative stress and the purine/adenosine system play a key role in their development and progression. Xanthine oxidase (XO) is a flavoprotein enzyme essential for the catalysis of the oxidative hydroxylation of purines -hypoxanthine and xanthine- to generate uric acid. As a consequence of the oxidative reaction of XO, reactive oxygen species (ROS) such as superoxide and hydrogen peroxide are produced and, further, contribute to the pathogenesis of mental disorders. Altered XO activity has been associated with free radical-mediated neurotoxicity inducing cell damage and inflammation. Diverse studies reported a direct association between an increased activity of XO and diverse mental diseases including depression or schizophrenia. Small-molecule inhibitors, such as the well-known allopurinol, and dietary flavonoids, can modulate the XO activity and subsequent ROS production. In the present work, we review the available literature on XO inhibition by small molecules and their potential therapeutic application in mental disorders. In addition, we discuss the chemistry and molecular mechanism of XO inhibitors, as well as the use of structure-based and computational methods to design specific inhibitors with the capability of modulating XO activity.
Collapse
Affiliation(s)
- Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepcion, 4070386 Concepcion, Chile
| | - Xavier Lucas
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel CH-4070, Switzerland
| | - Pedro Alarcón-Zapata
- Clinical Biochemistry and Immunology Department, Faculty of Pharmacy, University of Concepcion, 4070386 Concepcion, Chile
| | - Xavier Capó
- Research Group in Community Nutrition and Oxidative Stress, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), E-07122, Palma, Balearic Islands, Spain
| | - Maria Magdalena Quetglas-Llabrés
- Laboratory of Neurophysiology, Department of Biology, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), E-07122, Palma, Balearic Islands, Spain
| | - Silvia Tejada
- Laboratory of Neurophysiology, Department of Biology, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), E-07122, Palma, Balearic Islands, Spain
| | - Antoni Sureda
- Research Group in Community Nutrition and Oxidative Stress, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), E-07122, Palma, Balearic Islands, Spain
| |
Collapse
|
21
|
Guo Q, Zhao L, Zhu Y, Wu J, Hao C, Song S, Shi W. Optimization of culture medium for Sanghuangporus vaninii and a study on its therapeutic effects on gout. Biomed Pharmacother 2021; 135:111194. [PMID: 33395608 DOI: 10.1016/j.biopha.2020.111194] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/05/2020] [Accepted: 12/26/2020] [Indexed: 12/13/2022] Open
Abstract
The increasing incidence of gout poses a very challenging management problem. However, the currently available drugs often have various toxic side effects. As a traditional edible and medicinal macrofungus, Sanghuangporus vaninii presents high medical research value. Therefore, to improve fermentation efficiency and identify novel anti-gout drugs, we optimized the culture medium of S. vaninii with lignin and further investigated its anti-gout effects. The results indicated that 0.06 g/L of lignin was most favorable for S. vaninii growth. In the hyperuricemia cell model, we found that S. vaninii could significantly induce the downregulation of xanthine oxidoreductase and the upregulation of hypoxanthine-guanine phosphoribosyltransferase. Furthermore, following oral administration of the extracts, the serum uric acid levels of mice with hyperuricemia were effectively reduced. In a gouty arthritis rat model, S. vaninii also achieved strong suppression of synovial swelling, indicating its anti-inflammatory activity. In addition, the antioxidant assays suggested that S. vaninii shows a strong free radical scavenging capacity and can effectively alleviate cellular oxidative stress. This activity further enhances its anti-inflammatory activity and reduces the incidence of comorbidities. In summary, our results provide the basis for the utilization of S. vaninii to develop anti-gout drugs.
Collapse
Affiliation(s)
- Qiong Guo
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; College of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Liying Zhao
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; College of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Yuhua Zhu
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; College of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Jiang Wu
- School of Stomatology, Jiamusi University, Jiamusi, Heilongjiang, 154002, China
| | - Cuiting Hao
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; College of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Shuang Song
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; College of Life Sciences, Jilin University, Changchun, Jilin, 130012, China
| | - Wei Shi
- Key Laboratory for Molecular Enzymology & Engineering, The Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; College of Life Sciences, Jilin University, Changchun, Jilin, 130012, China.
| |
Collapse
|
22
|
Ju C, Lai RWC, Li KHC, Hung JKF, Lai JCL, Ho J, Liu Y, Tsoi MF, Liu T, Cheung BMY, Wong ICK, Tam LS, Tse G. Comparative cardiovascular risk in users versus non-users of xanthine oxidase inhibitors and febuxostat versus allopurinol users. Rheumatology (Oxford) 2021; 59:2340-2349. [PMID: 31873735 DOI: 10.1093/rheumatology/kez576] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/18/2019] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES The aim of this study is to determine major adverse cardiovascular events (MACE) and all-cause mortality comparing between xanthine oxidase inhibitors (XOIs) and non-XOI users, and between allopurinol and febuxostat. METHODS This is a retrospective cohort study of gout patients prescribed anti-hyperuricemic medications between 2013 and 2017 using a territory-wide administrative database. XOI users were matched 1:1 to XOI non-users using propensity scores. Febuxostat users were matched 1:3 to allopurinol users. Subgroup analyses were conducted based on colchicine use. RESULTS Of the 13 997 eligible participants, 3607 (25.8%) were XOI users and 10 390 (74.2%) were XOI non-users. After propensity score matching, compared with non-users (n = 3607), XOI users (n = 3607) showed similar incidence of MACE (hazard ratio [HR]: 0.997, 95% CI, 0.879, 1.131; P>0.05) and all-cause mortality (HR = 0.972, 95% CI 0.886, 1.065, P=0.539). Febuxostat (n = 276) users showed a similar risk of MACE compared with allopurinol users (n = 828; HR: 0.672, 95% CI, 0.416, 1.085; P=0.104) with a tendency towards a lower risk of heart failure-related hospitalizations (HR = 0.529, 95% CI 0.272, 1.029; P=0.061). Concurrent colchicine use reduced the risk for all-cause mortality amongst XOI users (HR = 0.671, 95% 0.586, 0.768; P<0.001). CONCLUSION In gout patients, XOI users showed similar risk of MACE and all-cause mortality compared with non-users. Compared with allopurinol users, febuxostat users showed similar MACE and all-cause mortality risks but lower heart failure-related hospitalizations.
Collapse
Affiliation(s)
- Chengsheng Ju
- School of Pharmacy, University College London, London, UK
| | - Rachel Wing Chuen Lai
- Laboratory of Cardiovascular Electrophysiology, Li Ka Shing Institute of Health Sciences, Hong Kong, P.R. China
| | | | - Joshua Kai Fung Hung
- Laboratory of Cardiovascular Electrophysiology, Li Ka Shing Institute of Health Sciences, Hong Kong, P.R. China
| | - Jenny C L Lai
- Department of Pharmacy & Pharmacology, University of Bath, Bath, UK
| | | | - Yingzhi Liu
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, Chinese University of Hong Kong
| | - Man Fung Tsoi
- Division of Clinical Pharmacology and Therapeutics, Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin
| | - Bernard Man Yung Cheung
- Division of Clinical Pharmacology and Therapeutics, Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ian Chi Kei Wong
- School of Pharmacy, University College London, London, UK
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong
| | - Lai Shan Tam
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, P.R. China
| |
Collapse
|
23
|
Cicero AFG, Fogacci F, Kuwabara M, Borghi C. Therapeutic Strategies for the Treatment of Chronic Hyperuricemia: An Evidence-Based Update. ACTA ACUST UNITED AC 2021; 57:medicina57010058. [PMID: 33435164 PMCID: PMC7827966 DOI: 10.3390/medicina57010058] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022]
Abstract
This article aims to critically review the evidence on the available therapeutic strategies for the treatment of hyperuricemia. For this reason, several papers were reviewed. Xanthine oxidase inhibitors are the safest and most effective uric acid lowering drugs for the management of chronic hyperuricemia, while the efficacy of uricosuric agents is strongly modulated by pharmacogenetics. Emergent drugs (lesinurad, peglotidase) were found to be more effective for the acute management of refractory hyperuricemia, but their use is supported by a relatively small number of clinical trials so that further well-designed clinical research is needed to deepen their efficacy and safety profile.
Collapse
Affiliation(s)
- Arrigo F. G. Cicero
- Hypertension Research Unit, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.F.G.C.); (F.F.)
| | - Federica Fogacci
- Hypertension Research Unit, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.F.G.C.); (F.F.)
| | - Masanari Kuwabara
- Cardiology Department and Intensive Care Unit, Toranomon Hospital, Tokyo 40138, Japan;
| | - Claudio Borghi
- Hypertension Research Unit, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (A.F.G.C.); (F.F.)
- Correspondence: ; Tel.: +39-512142224
| |
Collapse
|
24
|
Cicero AFG, Fogacci F, Cincione RI, Tocci G, Borghi C. Clinical Effects of Xanthine Oxidase Inhibitors in Hyperuricemic Patients. Med Princ Pract 2021; 30:122-130. [PMID: 33040063 PMCID: PMC8114083 DOI: 10.1159/000512178] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 10/07/2020] [Indexed: 12/13/2022] Open
Abstract
This review aims to critically present the available clinical evidence supporting the treatment of chronic hyperuricemia with xanthine oxidase inhibitors. For this reason, the studies published on uric acid (UA)-lowering drugs in the English language from 2000 to August 2019 have been carefully reviewed. The terms "serum uric acid," "xanthine oxidase," "allopurinol," "febuxostat," and "topiroxostat" were incorporated into an electronic search strategy, alone and in combinations, in both MEDLINE (National Library of Medicine, Bethesda, MD) and the Cochrane Register of Controlled Trials (The Cochrane Collaboration, Oxford, UK). Even if new urate-lowering drugs seem of particular efficacy for acute treatment of refractory hyperuricemia, their use is supported by relatively small clinical evidence. On the contrary, large long-term clinical trials have demonstrated that xanthine oxidase inhibitors (XOIs, namely, allopurinol and febuxostat) are effective, safe, and relatively well-tolerated in most of the patients. They have mainly been tested in the elderly, in patients affected by chronic diseases such as heart failure and cancer, and in patients taking a large number of drugs, confirming their safety profile. Recent data also show that they could exert some positive effects on vascular health, renal function, and glucose metabolism. Their cost is also low. In conclusion, XOIs remain the first choice of UA-lowering drug for chronic treatment.
Collapse
Affiliation(s)
- Arrigo F G Cicero
- Hypertension Research Unit, Medical and Surgical Sciences Department, University of Bologna, Bologna, Italy,
- IRCCS Policlinico di S.Orsola, Bologna, Italy,
| | - Federica Fogacci
- Hypertension Research Unit, Medical and Surgical Sciences Department, University of Bologna, Bologna, Italy
| | | | - Giuliano Tocci
- Hypertension Unit, Division of Cardiology, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, University of Rome Sapienza, Sant'Andrea Hospital, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Claudio Borghi
- Hypertension Research Unit, Medical and Surgical Sciences Department, University of Bologna, Bologna, Italy
- IRCCS Policlinico di S.Orsola, Bologna, Italy
| |
Collapse
|
25
|
Maresh MM, Abdelaziz RR, Ibrahim TM. Febuxostat mitigates concanavalin A-induced acute liver injury via modulation of MCP-1, IL-1β, TNF-α, neutrophil infiltration, and apoptosis in mice. Life Sci 2020; 260:118307. [PMID: 32841665 DOI: 10.1016/j.lfs.2020.118307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/10/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022]
Abstract
AIM Liver plays a crucial role in innate immunity reactions. This role predisposes the liver to innate-mediated liver injury when uncontrolled inflammation occurs. In this study, the effect of febuxostat administration on acute liver injury induced by concanavalin A (Con A) injection into mouse eye orbital sinus was studied. MATERIALS AND METHODS Two doses of febuxostat (10 and 20 mg/kg, orally) were administered either 1 h before or 30 min after the administration of Con A. Febuxostat at a low dose (10 mg/kg) before and after Con A modulated the elevation of serum ALT, liver uric acid, liver myeloperoxidase (MPO), and interleukin-1β (IL-1β) induced by Con A. The same dose of febuxostat before Con A also decreased serum total bilirubin and neutrophil infiltration, as evidenced by flow cytometry and histopathological analysis. KEY FINDINGS Febuxostat at a high dose (20 mg/kg) significantly improved serum ALT, AST, albumin, total bilirubin, liver uric acid, MPO, monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), interleukin-4 (IL-4), IL-1β, and neutrophil infiltration induced by Con A administration. The results of histopathological examination of liver cells paralleled the observed biochemical improvements. Hepatocyte apoptosis as evidenced by immunohistochemical examination of cleaved caspase-3 was markedly decreased in the febuxostat protection and treatment groups, in a dose-dependent manner SIGNIFICANCE: These results indicate that febuxostat, especially at the higher dose, may be an effective inhibitor of immune reactions evoked by Con A administration.
Collapse
Affiliation(s)
- Mohammed M Maresh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt..
| | - Tarek M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516 Mansoura, Egypt
| |
Collapse
|
26
|
Abstract
Xanthine oxidase inhibitors are primarily used in the clinical prevention and treatment of gout associated with hyperuricemia. The archetypal xanthine oxidase inhibitor, Allopurinol has been shown to have other beneficial effects such as a reduction in vascular reactive oxygen species and mechano-energetic uncoupling. This chapter discusses these properties and their relevance to human pathophysiology with a focus on Allopurinol as well as newer xanthine oxidase inhibitors such as Febuxostat and Topiroxostat. Xanthine oxidase (XO) and xanthine dehydrogenase (XDH) are collectively referred to as xanthine oxidoreductase (XOR). XDH is initially synthesised as a 150-kDa protein from which XO is derived, e.g. under conditions of ischemia/hypoxia either reversibly by conformational changes (calcium or SH oxidation) or irreversibly by proteolysis, the latter leading to formation of a 130-kDa form of XO. Both, XO and XDH, catalyse the conversion of hypoxanthine via xanthine to uric acid, the former by using oxygen forming superoxide and hydrogen peroxide and the latter NAD+. However, XDH is in principle also able to generate ROS.
Collapse
|
27
|
Pardue S, Kolluru GK, Shen X, Lewis SE, Saffle CB, Kelley EE, Kevil CG. Hydrogen sulfide stimulates xanthine oxidoreductase conversion to nitrite reductase and formation of NO. Redox Biol 2020; 34:101447. [PMID: 32035920 PMCID: PMC7327988 DOI: 10.1016/j.redox.2020.101447] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular disease is the leading cause of death and disability worldwide with increased oxidative stress and reduced NO bioavailability serving as key risk factors. For decades, elevation in protein abundance and enzymatic activity of xanthine oxidoreductase (XOR) under hypoxic/inflammatory conditions has been associated with organ damage and vascular dysfunction. Recent reports have challenged this dogma by identifying a beneficial function for XOR, under similar hypoxic/acidic conditions, whereby XOR catalyzes the reduction of nitrite (NO2-) to nitric oxide (NO) through poorly defined mechanisms. We previously reported that hydrogen sulfide (H2S/sulfide) confers significant vascular benefit under these same conditions via NO2- mediated mechanisms independent of nitric oxide synthase (NOS). Here we report for the first time the convergence of H2S, XOR, and nitrite to form a concerted triad for NO generation. Specifically, hypoxic endothelial cells show a dose-dependent, sulfide and polysulfide (diallyl trisulfide (DATS)-induced, NOS-independent NO2- reduction to NO that is dependent upon the enzymatic activity of XOR. Interestingly, nitrite reduction to NO was found to be slower and more sustained with DATS compared to H2S. Capacity for sulfide/polysulfide to produce an XOR-dependent impact on NO generation translates to salutary actions in vivo as DATS administration in cystathionine-γ-lyase (CSE) knockout mice significantly improved hindlimb ischemia blood flow post ligation, while the XOR-specific inhibitor, febuxostat (Febx), abrogated this benefit. Moreover, flow-mediated vasodilation (FMD) in CSE knockout mice following administration of DATS resulted in greater than 4-fold enhancement in femoral artery dilation while co-treatment with Febx completely completely abrogated this effect. Together, these results identify XOR as a focal point of convergence between sulfide- and nitrite-mediated signaling, as well as affirm the critical need to reexamine current dogma regarding inhibition of XOR in the context of vascular dysfunction.
Collapse
Affiliation(s)
- Sibile Pardue
- Department of Pathology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Gopi K Kolluru
- Department of Pathology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Xinggui Shen
- Department of Pathology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Sara E Lewis
- Department of Physiology and Pharmacology, West Virginia University, United States
| | - Courtney B Saffle
- Department of Physiology and Pharmacology, West Virginia University, United States
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University, United States
| | - Christopher G Kevil
- Department of Pathology, LSU Health Sciences Center, Shreveport, LA, USA; Department of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport, LA, USA; Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
28
|
Association between long-term prescription of febuxostat and the progression of heart failure with preserved ejection fraction in patients with hypertension and asymptomatic hyperuricemia. Heart Vessels 2020; 35:1446-1453. [DOI: 10.1007/s00380-020-01619-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 05/01/2020] [Indexed: 12/28/2022]
|
29
|
Tanaka A, Taguchi I, Teragawa H, Ishizaka N, Kanzaki Y, Tomiyama H, Sata M, Sezai A, Eguchi K, Kato T, Toyoda S, Ishibashi R, Kario K, Ishizu T, Ueda S, Maemura K, Higashi Y, Yamada H, Ohishi M, Yokote K, Murohara T, Oyama JI, Node K. Febuxostat does not delay progression of carotid atherosclerosis in patients with asymptomatic hyperuricemia: A randomized, controlled trial. PLoS Med 2020; 17:e1003095. [PMID: 32320401 PMCID: PMC7176100 DOI: 10.1371/journal.pmed.1003095] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND An elevated level of serum uric acid (SUA) is associated with an increased risk of cardiovascular disease. Pharmacological intervention with urate-lowering agents, such as the conventional purine analogue xanthine oxidase (XO) inhibitor, allopurinol, has been used widely for a long period of time in clinical practice to reduce SUA levels. Febuxostat, a novel non-purine selective inhibitor of XO, has higher potency for inhibition of XO activity and greater urate-lowering efficacy than conventional allopurinol. However, clinical evidence regarding the effects of febuxostat on atherosclerosis is lacking. The purpose of the study was to test whether treatment with febuxostat delays carotid intima-media thickness (IMT) progression in patients with asymptomatic hyperuricemia. METHODS AND FINDINGS The study was a multicenter, prospective, randomized, open-label, blinded-endpoint clinical trial undertaken at 48 sites throughout Japan between May 2014 and August 2018. Adults with both asymptomatic hyperuricemia (SUA >7.0 mg/dL) and maximum IMT of the common carotid artery (CCA) ≥1.1 mm at screening were allocated equally using a central web system to receive either dose-titrated febuxostat (10-60 mg daily) or as a control-arm, non-pharmacological lifestyle modification for hyperuricemia, such as a healthy diet and exercise therapy. Of the 514 enrolled participants, 31 were excluded from the analysis, with the remaining 483 people (mean age 69.1 years [standard deviation 10.4 years], female 19.7%) included in the primary analysis (febuxostat group, 239; control group, 244), based on a modified intention-to-treat principal. The carotid IMT images were recorded by a single sonographer at each site and read in a treatment-blinded manner by a single analyzer at a central core laboratory. The primary endpoint was the percentage change from baseline to 24 months in mean IMT of the CCA, determined by analysis of covariance using the allocation adjustment factors (age, gender, history of type 2 diabetes, baseline SUA, and baseline maximum IMT of the CCA) as the covariates. Key secondary endpoints included changes in other carotid ultrasonographic parameters and SUA and the incidence of clinical events. The mean values (± standard deviation) of CCA-IMT were 0.825 mm ± 0.173 mm in the febuxostat group and 0.832 mm ± 0.175 mm in the control group (mean between-group difference [febuxostat - control], -0.007 mm [95% confidence interval (CI) -0.039 mm to 0.024 mm; P = 0.65]) at baseline; 0.832 mm ± 0.182 mm in the febuxostat group and 0.848 mm ± 0.176 mm in the control group (mean between-group difference, -0.016 mm [95% CI -0.051 mm to 0.019 mm; P = 0.37]) at 24 months. Compared with the control group, febuxostat had no significant effect on the primary endpoint (mean percentage change 1.2% [95% CI -0.6% to 3.0%] in the febuxostat group (n = 207) versus 1.4% [95% CI -0.5% to 3.3%] in the control group (n = 193); mean between-group difference, -0.2% [95% CI -2.3% to 1.9%; P = 0.83]). Febuxostat also had no effect on the other carotid ultrasonographic parameters. The mean baseline values of SUA were comparable between the two groups (febuxostat, 7.76 mg/dL ± 0.98 mg/dL versus control, 7.73 mg/dL ± 1.04 mg/dL; mean between-group difference, 0.03 mg/dL [95% CI -0.15 mg/dL to 0.21 mg/dL; P = 0.75]). The mean value of SUA at 24 months was significantly lower in the febuxostat group than in the control group (febuxostat, 4.66 mg/dL ± 1.27 mg/dL versus control, 7.28 mg/dL ± 1.27 mg/dL; mean between-group difference, -2.62 mg/dL [95% CI -2.86 mg/dL to -2.38 mg/dL; P < 0.001]). Episodes of gout arthritis occurred only in the control group (4 patients [1.6%]). There were three deaths in the febuxostat group and seven in the control group during follow-up. A limitation of the study was the study design, as it was not a placebo-controlled trial, had a relatively small sample size and a short intervention period, and only enrolled Japanese patients with asymptomatic hyperuricemia. CONCLUSIONS In Japanese patients with asymptomatic hyperuricemia, 24 months of febuxostat treatment did not delay carotid atherosclerosis progression, compared with non-pharmacological care. These findings do not support the use of febuxostat for delaying carotid atherosclerosis in this population. TRIAL REGISTRATION University Hospital Medical Information Network Clinical Trial Registry UMIN000012911.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
- * E-mail: (AT); (KN)
| | - Isao Taguchi
- Department of Cardiology, Dokkyo Medical University Saitama Medical Center, Koshigaya, Japan
| | - Hiroki Teragawa
- Department of Cardiovascular Medicine, JR Hiroshima Hospital, Hiroshima, Japan
| | | | - Yumiko Kanzaki
- Department of Cardiology, Osaka Medical College, Takatsuki, Japan
| | | | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Akira Sezai
- The Department of Cardiovascular Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Kazuo Eguchi
- Department of Internal Medicine, Hanyu General Hospital, Hanyu, Japan
| | - Toru Kato
- Department of Clinical Research, National Hospital Organization, Tochigi Medical Center, Utsunomiya, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University, Mibu, Japan
| | - Ryoichi Ishibashi
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Kimitsu Chuo Hospital, Kisarazu, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Tomoko Ishizu
- Department of Clinical Laboratory Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shinichiro Ueda
- Department of Clinical Pharmacology and Therapeutics, University of the Ryukyus, Nishihara, Japan
| | - Koji Maemura
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Hirotsugu Yamada
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Mitsuru Ohishi
- Department of Cardiovascular Medical and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kotaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun-ichi Oyama
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
- * E-mail: (AT); (KN)
| | | |
Collapse
|
30
|
Zhang S, Wang Y, Cheng J, Huangfu N, Zhao R, Xu Z, Zhang F, Zheng W, Zhang D. Hyperuricemia and Cardiovascular Disease. Curr Pharm Des 2020; 25:700-709. [PMID: 30961478 DOI: 10.2174/1381612825666190408122557] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/31/2019] [Indexed: 02/07/2023]
Abstract
Purine metabolism in the circulatory system yields uric acid as its final oxidation product, which is believed to be linked to the development of gout and kidney stones. Hyperuricemia is closely correlated with cardiovascular disease, metabolic syndrome, and chronic kidney disease, as attested by the epidemiological and empirical research. In this review, we summarize the recent knowledge about hyperuricemia, with a special focus on its physiology, epidemiology, and correlation with cardiovascular disease. This review also discusses the possible positive effects of treatment to reduce urate levels in patients with cardiovascular disease and hyperuricemia, which may lead to an improved clinical treatment plan.
Collapse
Affiliation(s)
- Shuangshuang Zhang
- Department of Cardiovascular Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315000, China
| | - Yong Wang
- Department of Cardiovascular Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315000, China
| | - Jinsong Cheng
- Department of Cardiovascular Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315000, China
| | - Ning Huangfu
- Department of Cardiovascular Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315000, China
| | - Ruochi Zhao
- Department of Cardiovascular Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315000, China
| | - Zhenyu Xu
- Department of Cardiovascular Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315000, China
| | - Fuxing Zhang
- Department of Cardiovascular Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315000, China
| | - Wenyuan Zheng
- Department of Cardiovascular Medicine, Ningbo First Hospital, Ningbo, Zhejiang 315000, China
| | - Dandan Zhang
- Department of Pharmacy, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
31
|
Albu A, Para I, Porojan M. Uric Acid and Arterial Stiffness. Ther Clin Risk Manag 2020; 16:39-54. [PMID: 32095074 PMCID: PMC6995306 DOI: 10.2147/tcrm.s232033] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 12/01/2019] [Indexed: 12/21/2022] Open
Abstract
Hyperuricemia is usually associated with hypertension, diabetes mellitus, metabolic syndrome and chronic kidney disease. Accumulating data from epidemiological studies indicate an association of increased uric acid (UA) with cardiovascular diseases. Possible pathogenic mechanisms include enhancement of oxidative stress and systemic inflammation caused by hyperuricemia. Arterial stiffness may be one of the possible pathways between hyperuricemia and cardiovascular disease, but a clear relationship between increased UA and vascular alterations has not been confirmed. The review summarizes the epidemiological studies investigating the relationship between UA and arterial stiffness and highlights the results of interventional studies evaluating arterial stiffness parameters in patients treated with UA-lowering drugs.
Collapse
Affiliation(s)
| | - Ioana Para
- 4th Department of Internal Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | |
Collapse
|
32
|
El-Sheikh AA, Abdelzaher WY, Gad AA, Abdel-Gaber SA. Purine versus non-purine xanthine oxidase inhibitors against cyclophosphamide-induced cardiac and bone marrow toxicity in rats. Hum Exp Toxicol 2019; 39:249-261. [PMID: 31640406 DOI: 10.1177/0960327119883412] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIM Cancer is a fatal and serious disease. Cyclophosphamide (CYC) is a commonly used anticancer drug. Cardiotoxicity and myelotoxicity are life-threatening side effects of CYC treatment. We aimed to evaluate the effect of the xanthine oxidase (XO) inhibitors, allopurinol (ALL) and febuxostat (FEB), on CYC-induced cardio- and hematopoietic toxicity in rats. METHODS ALL (100 mg/kg/day) or FEB (10 mg/kg/day) were administered orally to rats in the presence and absence of CYC (200 mg/kg kg i.p. single dose) treatment. Serum creatine kinase-MB creatine kinase myocardial band (CK-MB) and lactate dehydrogenase (LDH) activities were estimated. Complete blood counting (CBC), cardiac and bone marrow XO activity, malondialdehyde level, and superoxide dismutase activity were determined. Cardiac and bone marrow histopathological changes were also evaluated. RESULTS ALL and FEB significantly decreased CK-MB and LDH induced by CYC. Disturbed levels of XO, oxidative stress parameters, and CBC were also corrected by both XO inhibitors tested, with amelioration of cardiac histopathological changes caused by CYC. Treatment with FEB, but not ALL, prior to CYC challenges normalized bone marrow histopathological changes. CONCLUSION These results suggest that both XO inhibitors tested; ALL and FEB can ameliorate CYC-induced cardiotoxicity. However, only FEB can protect against CYC-induced myelotoxicity, whereas ALL, to the contrary, might aggravate it.
Collapse
Affiliation(s)
- A A El-Sheikh
- Basic Health Sciences Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.,Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - W Y Abdelzaher
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - A A Gad
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - S A Abdel-Gaber
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
33
|
Brombo G, Bonetti F, Volpato S, Morieri ML, Napoli E, Bandinelli S, Cherubini A, Maggio M, Guralnik J, Ferrucci L, Zuliani G. Uric acid within the "normal" range predict 9-year cardiovascular mortality in older individuals. The InCHIANTI study. Nutr Metab Cardiovasc Dis 2019; 29:1061-1067. [PMID: 31377184 PMCID: PMC6744329 DOI: 10.1016/j.numecd.2019.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Increased uric acid levels correlate with cardiovascular disease and cardiovascular/overall mortality. To identify a uric acid threshold above which cardiovascular mortality rises, we studied the relationship between uric acid concentration and overall/cardiovascular mortality. METHODS AND RESULTS We analyzed data from the InCHIANTI study, a cohort study of Italian community-dwelling people with 9 years of follow-up. We selected a sample of 947 individuals over 64 years of age, free from cardio-cerebrovascular disease and with available uric acid measurement at baseline. The sample was divided according to plasma uric acid tertiles. The Hazard ratio (HR) for mortality was calculated by multivariate Cox proportional hazard model. Mean age of participants was 75.3 ± 7.3 years; the mean value of uric acid was 5.1 ± 1.4 mg/dl. Over 9-years of follow-up, 342 (36.1%) participants died, 143 deaths (15.1%) were due to cardiovascular disease. Subjects with higher uric acid concentrations presented a higher cardiovascular mortality [II (4.6-5.5 mg/dl) vs I (1.8-4.5 mg/dl) tertile HR: 1.98, 95%C.I. 1.22-3.23; III (≥5.6 mg/dl) vs I tertile HR: 1.87, 95%C.I. 1.13-3.09]. We found a non-linear association between uric acid concentrations and cardiovascular mortality with the lowest mortality for values of about 4.1 mg/dl and a significant risk increment for values above 4.3 mg/dl. CONCLUSION In community-dwelling older individuals free from cardio-cerebrovascular events, the lowest 9-year cardiovascular mortality was observed for uric acid values far below current target values. If confirmed, these data might represent the background for investigating the efficacy of uric acid levels reduction in similar populations.
Collapse
Affiliation(s)
- Gloria Brombo
- Department of Medical Sciences, Section of Internal and Cardiorespiratory Medicine, University of Ferrara, Italy.
| | - Francesco Bonetti
- Department of Medical Sciences, Section of Internal and Cardiorespiratory Medicine, University of Ferrara, Italy
| | - Stefano Volpato
- Department of Medical Sciences, Section of Internal and Cardiorespiratory Medicine, University of Ferrara, Italy
| | - Mario L Morieri
- Department of Medical Sciences, Section of Internal and Cardiorespiratory Medicine, University of Ferrara, Italy
| | - Ettore Napoli
- Department of Medical Sciences, Section of Internal and Cardiorespiratory Medicine, University of Ferrara, Italy
| | - Stefania Bandinelli
- Geriatric Rehabilitation Unit, Tuscany Regional Health Agency, Florence, Italy
| | | | - Marcello Maggio
- Department of Clinical and Experimental Medicine, Section of Geriatrics, University of Parma, Italy
| | - Jack Guralnik
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Clinical Research Branch, National Institute on Aging, NIH, Baltimore, USA
| | - Giovanni Zuliani
- Department of Morphology, Surgery and Experimental Medicine, Section of Internal and Cardiorespiratory Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
34
|
Electrochemical investigation of the inhibition effect of carvacrol on xanthine oxidase activity merging with theoretical studies. Process Biochem 2019. [DOI: 10.1016/j.procbio.2019.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
35
|
Adusumalli S, Jamwal R, Obach RS, Ryder TF, Leggio L, Akhlaghi F. Role of Molybdenum-Containing Enzymes in the Biotransformation of the Novel Ghrelin Receptor Inverse Agonist PF-5190457: A Reverse Translational Bed-to-Bench Approach. Drug Metab Dispos 2019; 47:874-882. [PMID: 31182423 DOI: 10.1124/dmd.119.087015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/28/2019] [Indexed: 12/29/2022] Open
Abstract
(R)-2-(2-methylimidazo[2,1-b]thiazol-6-yl)-1-(2-(5-(6-methylpyrimidin-4-yl)-2,3-dihydro-1H-inden-1-yl)-2,7-diazaspiro[3.5]nonan-7-yl)ethan-1-one (PF-5190457) was identified as a potent and selective inverse agonist of the ghrelin receptor [growth hormone secretagogue receptor 1a (GHS-R1a)]. The present translational bed-to-bench work characterizes the biotransformation of this compound in vivo and then further explores in vitro metabolism in fractions of human liver and primary hepatocytes. Following oral administration of PF-5190457 in a phase 1b clinical study, hydroxyl metabolites of the compound were observed, including one that had not been observed in previously performed human liver microsomal incubations. PF-6870961 was biosynthesized using liver cytosol, and the site of hydroxylation was shown to be on the pyrimidine using nuclear magnetic resonance spectroscopy. The aldehyde oxidase (AO) inhibitor raloxifene and the xanthine oxidase inhibitor febuxostat inhibited the formation of PF-6870961 in human liver cytosol, suggesting both enzymes were involved in the metabolism of the drug. However, greater inhibition was observed with raloxifene, indicating AO is a dominant enzyme in the biotransformation. The intrinsic clearance of the drug in human liver cytosol was estimated to be 0.002 ml/min per milligram protein. This study provides important novel information at three levels: 1) it provides additional new information on the recently developed novel compound PF-5190457, the first GHS-R1a blocker that has moved to development in humans; 2) it provides an example of a reverse translational approach where a discovery in humans was brought back, validated, and further investigated at the bench level; and 3) it demonstrates the importance of considering the molybdenum-containing oxidases during the development of new drug entities. SIGNIFICANCE STATEMENT: PF-5190457 is a novel ghrelin receptor inverse agonist that is currently undergoing clinical development for treatment of alcohol use disorder. PF-6870961, a major hydroxyl metabolite of the compound, was observed in human plasma, but was absent in human liver microsomal incubations. PF-6870961 was biosynthesized using liver cytosol, and the site of hydroxylation on the pyrimidine ring was characterized. Inhibitors of aldehyde oxidase and xanthine oxidase inhibited the formation of PF-6870961 in human liver cytosol, suggesting both enzymes were involved in the metabolism of the drug. This information is important for patient selection in subsequent clinical studies.
Collapse
Affiliation(s)
- Sravani Adusumalli
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - Rohitash Jamwal
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - R Scott Obach
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - Tim F Ryder
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - Lorenzo Leggio
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., R.J., F.A.); Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut (R.S.O., T.F.R.); Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research and National Institute on Drug Abuse Intramural Research Program, Bethesda, Maryland (L.L.); Medication Development Program, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (L.L.); and Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island (L.L.)
| |
Collapse
|
36
|
Li F, Dong YZ, Zhang D, Zhang XM, Lin ZJ, Zhang B. Molecular mechanisms involved in drug-induced liver injury caused by urate-lowering Chinese herbs: A network pharmacology study and biology experiments. PLoS One 2019; 14:e0216948. [PMID: 31141540 PMCID: PMC6541264 DOI: 10.1371/journal.pone.0216948] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/01/2019] [Indexed: 12/12/2022] Open
Abstract
As an important part of the comprehensive treatment methods, the urate-lowering Chinese herbs could provide favorable clinical effects on hyperuricemia in its ability to invigorate spleen and remove dampness. Owing to the long-term duration, it brought up the potential adverse reactions (ADRs) and concerns about the drug-induced liver injury from these herbs. To address this problem, the bioinformatics approaches which combined the network pharmacology, computer simulation and molecular biology experiments were undertaken to elucidate the underlying drug-induced liver injury molecular mechanisms of urate-lowering Chinese herbs. Several electronic databases were searched to identify the potential liver injury compounds in published research. Then, the putative target profile of liver injury was predicted, and the interaction network was constructed based on the links between the compounds, corresponding targets and core pathways. Accordingly, the molecular docking simulation was performed to recognize the representative compounds with hepatotoxicity. Finally, the cell experiments were conducted to investigate the biochemical indicators and expression of the crucial protein that were closely associated with liver injury. In conclusion, the current research revealed that the compounds with potential liver injury including diosgenin, baicalin, saikosaponin D, tetrandrine, rutaecarpine and evodiamine from urate-lowering Chinese herbs, could lead to decline the survival rate of L-02 cell, increase the activities of aspartate aminotransferase (AST), alanine aminotransferase (ALT), lactate dehydrogenase (LDH) and alkaline phosphatase (ALP) in cell-culture medium, enhance the expression of p-p38/p38, while the p38 inhibitor could achieve the trend of regulating and controlling liver injury. These research findings bring further support to the growing evidence that the mechanism of the liver injury induced by the compounds from urate-lowering Chinese herbs may be associated with the activation of p38α.
Collapse
Affiliation(s)
- Fan Li
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Chao Yang District, Beijing, China
| | - Yi-Zhu Dong
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Chao Yang District, Beijing, China
| | - Dan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Chao Yang District, Beijing, China
| | - Xiao-Meng Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Chao Yang District, Beijing, China
| | - Zhi-Jian Lin
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Chao Yang District, Beijing, China
| | - Bing Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Chao Yang District, Beijing, China
- * E-mail:
| |
Collapse
|
37
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
38
|
Deciphering the inhibition effect of thymoquinone on xanthine oxidase activity using differential pulse voltammetry in combination with theoretical studies. Enzyme Microb Technol 2019; 121:29-36. [DOI: 10.1016/j.enzmictec.2018.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 02/07/2023]
|
39
|
Kelley EE. Diminishing Inflammation by Reducing Oxidant Generation: Nitrated Fatty Acid-Mediated Inactivation of Xanthine Oxidoreductase. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1127:59-65. [PMID: 31140171 DOI: 10.1007/978-3-030-11488-6_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Inhibition of xanthine oxidoreductase (XOR) has proven beneficial in a plethora of inflammatory disease processes due to a net reduction in pro-inflammatory oxidants and secondary nitrating species. Electrophilic nitrated fatty acid derivatives, such as nitro-oleic acid (OA-NO2) are also noted to display a broad spectrum of anti-inflammatory effects via interaction with critical signaling pathways. An alternative process in which nitrated fatty acids may extend anti-inflammatory actions is via inactivation of XOR, a process that is more effective than allo/oxypurinol-mediated inhibition. Herein, we describe the molecular aspects of nitrated fatty acid-associated inactivation of XOR, identify specificity via structure function relationships and discuss XOR as a crucial component of the anti-inflammatory portfolio of nitrated fatty acids.
Collapse
Affiliation(s)
- Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
40
|
The impact of xanthine oxidase (XO) on hemolytic diseases. Redox Biol 2018; 21:101072. [PMID: 30580157 PMCID: PMC6305892 DOI: 10.1016/j.redox.2018.101072] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 01/13/2023] Open
Abstract
Hemolytic diseases are associated with elevated levels of circulating free heme that can mediate endothelial dysfunction directly via redox reactions with biomolecules or indirectly by upregulating enzymatic sources of reactive species. A key enzymatic source of these reactive species is the purine catabolizing enzyme, xanthine oxidase (XO) as the oxidation of hypoxanthine to xanthine and subsequent oxidation of xanthine to uric acid generates superoxide (O2•-) and hydrogen peroxide (H2O2). While XO has been studied for over 120 years, much remains unknown regarding specific mechanistic roles for this enzyme in pathologic processes. This gap in knowledge stems from several interrelated issues including: 1) lethality of global XO deletion and the absence of tissue-specific XO knockout models have coalesced to relegate proof-of-principle experimentation to pharmacology; 2) XO is mobile and thus when upregulated locally can be secreted into the circulation and impact distal vascular beds by high-affinity association to the glycocalyx on the endothelium; and 3) endothelial-bound XO is significantly resistant (> 50%) to inhibition by allopurinol, the principle compound used for XO inhibition in the clinic as well as the laboratory. While it is known that circulating XO is elevated in hemolytic diseases including sickle cell, malaria and sepsis, little is understood regarding its role in these pathologies. As such, the aim of this review is to define our current understanding regarding the effect of hemolysis (free heme) on circulating XO levels as well as the subsequent impact of XO-derived oxidants in hemolytic disease processes.
Collapse
|
41
|
An in-depth view of potential dual effect of thymol in inhibiting xanthine oxidase activity: Electrochemical measurements in combination with four way PARAFAC analysis and molecular docking insights. Int J Biol Macromol 2018; 119:1298-1310. [DOI: 10.1016/j.ijbiomac.2018.08.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/25/2018] [Accepted: 08/05/2018] [Indexed: 02/01/2023]
|
42
|
Costa JDS, Ramos RDS, Costa KDSL, Brasil DDSB, Silva CHTDPD, Ferreira EFB, Borges RDS, Campos JM, Macêdo WJDC, Santos CBRD. An In Silico Study of the Antioxidant Ability for Two Caffeine Analogs Using Molecular Docking and Quantum Chemical Methods. Molecules 2018; 23:molecules23112801. [PMID: 30380600 PMCID: PMC6278550 DOI: 10.3390/molecules23112801] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 10/01/2018] [Accepted: 10/11/2018] [Indexed: 12/13/2022] Open
Abstract
The antioxidant activity of molecules constitutes an important factor for the regulation of redox homeostasis and reduction of the oxidative stress. Cells affected by oxidative stress can undergo genetic alteration, causing structural changes and promoting the onset of chronic diseases, such as cancer. We have performed an in silico study to evaluate the antioxidant potential of two molecules of the zinc database: ZINC08706191 (Z91) and ZINC08992920 (Z20). Molecular docking, quantum chemical calculations (HF/6-31G**) and Pearson’s correlation have been performed. Molecular docking results of Z91 and Z20 showed both the lower binding affinity (BA) and inhibition constant (Ki) values for the receptor-ligand interactions in the three tested enzymes (cytochrome P450—CP450, myeloperoxidase—MP and NADPH oxidase—NO) than the control molecules (5-fluorouracil—FLU, melatonin—MEL and dextromethorphan—DEX, for each receptor respectively). Molecular descriptors were correlated with Ki and strong correlations were observed for the CP450, MP and NO receptors. These and other results attest the significant antioxidant ability of Z91 and Z20, that may be indicated for further analyses in relation to the control of oxidative stress and as possible antioxidant agents to be used in the pharmaceutical industry.
Collapse
Affiliation(s)
- Josivan da Silva Costa
- Postgraduate Program in Biotechnology and Biodiversity-Network BIONORTE, Federal University of Pará, Rua Augusto Corrêa, 01, Belém, Pará 66075110, Brazil.
- Laboratory of Modeling and Computational Chemistry, Department of Biological Sciences, Federal University of Amapá, Rod. Juscelino Kubitschek, Km 02, s/n, Macapá, Amapá 68902-280, Brazil.
- Laboratory of Molecular Modeling and Simulation System, Federal Rural University of Amazônia, Rua João Pessoa, 121, Capanema, Pará 68700-030, Brazil.
| | - Ryan da Silva Ramos
- Laboratory of Modeling and Computational Chemistry, Department of Biological Sciences, Federal University of Amapá, Rod. Juscelino Kubitschek, Km 02, s/n, Macapá, Amapá 68902-280, Brazil.
- Laboratory of Molecular Modeling and Simulation System, Federal Rural University of Amazônia, Rua João Pessoa, 121, Capanema, Pará 68700-030, Brazil.
| | - Karina da Silva Lopes Costa
- Laboratory of Modeling and Computational Chemistry, Department of Biological Sciences, Federal University of Amapá, Rod. Juscelino Kubitschek, Km 02, s/n, Macapá, Amapá 68902-280, Brazil.
| | | | | | - Elenilze Figueiredo Batista Ferreira
- Laboratory of Modeling and Computational Chemistry, Department of Biological Sciences, Federal University of Amapá, Rod. Juscelino Kubitschek, Km 02, s/n, Macapá, Amapá 68902-280, Brazil.
| | - Rosivaldo Dos Santos Borges
- Postgraduate Program in Biotechnology and Biodiversity-Network BIONORTE, Federal University of Pará, Rua Augusto Corrêa, 01, Belém, Pará 66075110, Brazil.
| | - Joaquín María Campos
- Department of Pharmaceutical and Organic Chemistry, University of Granada, Campus of Cartuja, 18071 Granada, Spain.
| | - Williams Jorge da Cruz Macêdo
- Postgraduate Program in Biotechnology and Biodiversity-Network BIONORTE, Federal University of Pará, Rua Augusto Corrêa, 01, Belém, Pará 66075110, Brazil.
- Laboratory of Molecular Modeling and Simulation System, Federal Rural University of Amazônia, Rua João Pessoa, 121, Capanema, Pará 68700-030, Brazil.
| | - Cleydson Breno Rodrigues Dos Santos
- Laboratory of Modeling and Computational Chemistry, Department of Biological Sciences, Federal University of Amapá, Rod. Juscelino Kubitschek, Km 02, s/n, Macapá, Amapá 68902-280, Brazil.
- Laboratory of Molecular Modeling and Simulation System, Federal Rural University of Amazônia, Rua João Pessoa, 121, Capanema, Pará 68700-030, Brazil.
- Department of Pharmaceutical and Organic Chemistry, University of Granada, Campus of Cartuja, 18071 Granada, Spain.
| |
Collapse
|
43
|
Yokota T, Fukushima A, Kinugawa S, Okumura T, Murohara T, Tsutsui H. Randomized Trial of Effect of Urate-Lowering Agent Febuxostat in Chronic Heart Failure Patients with Hyperuricemia (LEAF-CHF). Int Heart J 2018; 59:976-982. [PMID: 30101851 DOI: 10.1536/ihj.17-560] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Hyperuricemia is an independent predictor of mortality in patients with chronic heart failure. The aim of the study is to determine whether a urate-lowering agent febuxostat, an inhibitor of xanthine oxidase, may improve the clinical outcomes in chronic heart failure patients with hyperuricemia when compared to conventional treatment. This multicenter, prospective, randomized, open-label, blinded endpoint study with a follow-up period of 24 weeks will enroll 200 Japanese chronic heart failure patients with hyperuricemia. The eligibility criteria include a diagnosis of chronic heart failure (New York Heart Association functional class II-III with a history of hospitalization due to worsening of heart failure within the last 2 years), reduced left ventricular systolic function (left ventricular ejection fraction < 40%) and increased plasma natriuretic peptide [plasma B-type natriuretic peptide (BNP) ≥ 100 pg/mL or N-terminal pro BNP (NT-proBNP) ≥ 400 pg/mL], and hyperuricemia (serum uric acid >7.0 mg/dL and ≤ 10 mg/dL) at the screening visit. The primary outcome is the difference in the plasma BNP levels between the baseline and 24 weeks of treatment. The plasma BNP levels are measured in the central laboratory in a blinded manner. This study investigates the efficacy and safety of febuxostat in chronic heart failure patients with hyperuricemia.
Collapse
Affiliation(s)
- Takashi Yokota
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Arata Fukushima
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Hiroyuki Tsutsui
- Department Cardiovascular Medicine, Faculty of Medical Sciences, Kyusyu University
| |
Collapse
|
44
|
An Antioxidant Potential, Quantum-Chemical and Molecular Docking Study of the Major Chemical Constituents Present in the Leaves of Curatella americana Linn. Pharmaceuticals (Basel) 2018; 11:ph11030072. [PMID: 30036950 PMCID: PMC6161303 DOI: 10.3390/ph11030072] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are continuously generated in the normal biological systems, primarily by enzymes as xanthine oxidase (XO). The inappropriate scavenging or inhibition of ROS has been considered to be linked with aging, inflammatory disorders, and chronic diseases. Therefore, many plants and their products have been investigated as natural antioxidants for their potential use in preventive medicine. The leaves and bark extracts of Curatella americana Linn. were described in scientific research as anti-inflammatory, vasodilator, anti-ulcerogenic, and hypolipidemic effects. So, the aim of this study was to evaluate the antioxidant potentials of leaf hydroalcoholic extract from C. americana (HECA) through the scavenging DPPH assay and their main chemical constituents, evaluated by the following quantum chemical approaches (DFT B3LYP/6-31G**): Maps of Molecular Electrostatic Potential (MEP), Frontier Orbital’s (HOMO and LUMO) followed by multivariate analysis and molecular docking simulations with the xanthine oxidase enzyme. The hydroalcoholic extract showed significant antioxidant activity by free radical scavenging probably due to the great presence of flavonoids, which were grouped in the PCA and HCA analysis with the standard gallic acid. In the molecular docking study, the compounds studied presented the binding free energy (ΔG) values close each other, due to the similar interactions with amino acids residues at the activity site. The descriptors Gap and softness were important to characterize the molecules with antioxidant potential by capturing oxygen radicals.
Collapse
|
45
|
Liu X, Li Y, Li Z, Wei X, Ma Y, Cheng P, Jiao R, Fang J, Xing Y, Tang J, Wang M, Li T. A novel IgG1 monoclonal antibody against xanthine oxidase alleviates inflammation induced by potassium oxonate in mice. Int J Biol Macromol 2018; 112:537-547. [DOI: 10.1016/j.ijbiomac.2018.01.171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 11/29/2022]
|
46
|
Role of Hyperhomocysteinemia and Hyperuricemia in Pathogenesis of Atherosclerosis. J Stroke Cerebrovasc Dis 2017; 26:2695-2699. [DOI: 10.1016/j.jstrokecerebrovasdis.2016.10.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/01/2016] [Accepted: 10/10/2016] [Indexed: 11/17/2022] Open
|
47
|
Abstract
BACKGROUNDS Although evidence is mounting on the role of hyperuricemia in cardio-renal disease, continuing doubt remains as to whether hyperuricemia can be considered a major causal cardiovascular risk factor. In addition, available data suggest that treatment may be beneficial, even in the absence of overt gout, when hyperuricemia accompanies other clinical conditions, such as urate deposition, advanced chronic kidney disease, or cardiovascular risk factors. METHODS AND RESULTS Analysis of the literature suggests there would be sufficient evidence warranting clinical trials to determine whether lowering uric acid levels would be clinically beneficial in the prevention or treatment of cardiovascular and renal diseases. CONCLUSION Under a practical profile, it becomes progressively more important to investigate the possibility of reducing serum uric acid levels in the general population below the level of 5.0 mg/dL.
Collapse
Affiliation(s)
- Claudio Ferri
- a Division of Internal Medicine and Nephrology, School of Internal Medicine , University of L'Aquila, San Salvatore Hospital Coppito , L'Aquila , Italy
| |
Collapse
|
48
|
Affiliation(s)
- Claudio Borghi
- a Department of Medical and Surgical Science (DIMEC) , Alma Mater Studiorum University of Bologna , Bologna , Italy
| |
Collapse
|
49
|
Xanthine oxidoreductase and its inhibitors: relevance for gout. Clin Sci (Lond) 2017; 130:2167-2180. [PMID: 27798228 DOI: 10.1042/cs20160010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 09/14/2016] [Indexed: 12/22/2022]
Abstract
Xanthine oxidoreductase (XOR) is the rate-limiting enzyme in purine catabolism and converts hypoxanthine to xanthine, and xanthine into uric acid. When concentrations of uric acid exceed its biochemical saturation point, crystals of uric acid, in the form of monosodium urate, emerge and can predispose an individual to gout, the commonest form of inflammatory arthritis in men aged over 40 years. XOR inhibitors are primarily used in the treatment of gout, reducing the formation of uric acid and thereby, preventing the formation of monosodium urate crystals. Allopurinol is established as first-line therapy for gout; a newer alternative, febuxostat, is used in patients unable to tolerate allopurinol. This review provides an overview of gout, a detailed analysis of the structure and function of XOR, discussion on the pharmacokinetics and pharmacodynamics of XOR inhibitors-allopurinol and febuxostat, and the relevance of XOR in common comorbidities of gout.
Collapse
|
50
|
Zimmerman MC, Clemens DL, Duryee MJ, Sarmiento C, Chiou A, Hunter CD, Tian J, Klassen LW, O'Dell JR, Thiele GM, Mikuls TR, Anderson DR. Direct antioxidant properties of methotrexate: Inhibition of malondialdehyde-acetaldehyde-protein adduct formation and superoxide scavenging. Redox Biol 2017; 13:588-593. [PMID: 28803127 PMCID: PMC5552384 DOI: 10.1016/j.redox.2017.07.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 07/26/2017] [Accepted: 07/29/2017] [Indexed: 12/14/2022] Open
Abstract
Methotrexate (MTX) is an immunosuppressant commonly used for the treatment of autoimmune diseases. Recent observations have shown that patients treated with MTX also exhibit a reduced risk for the development of cardiovascular disease (CVD). Although MTX reduces systemic inflammation and tissue damage, the mechanisms by which MTX exerts these beneficial effects are not entirely known. We have previously demonstrated that protein adducts formed by the interaction of malondialdehyde (MDA) and acetaldehyde (AA), known as MAA-protein adducts, are present in diseased tissues of individuals with rheumatoid arthritis (RA) or CVD. In previously reported studies, MAA-adducts were shown to be highly immunogenic, supporting the concept that MAA-adducts not only serve as markers of oxidative stress but may have a direct role in the pathogenesis of inflammatory diseases. Because MAA-adducts are commonly detected in diseased tissues and are proposed to mitigate disease progression in both RA and CVD, we tested the hypothesis that MTX inhibits the generation of MAA-protein adducts by scavenging reactive oxygen species. Using a cell free system, we found that MTX reduces MAA-adduct formation by approximately 6-fold, and scavenges free radicals produced during MAA-adduct formation. Further investigation revealed that MTX directly scavenges superoxide, but not hydrogen peroxide. Additionally, using the Nrf2/ARE luciferase reporter cell line, which responds to intracellular redox changes, we observed that MTX inhibits the activation of Nrf2 in cells treated with MDA and AA. These studies define previously unrecognized mechanisms by which MTX can reduce inflammation and subsequent tissue damage, namely, scavenging free radicals, reducing oxidative stress, and inhibiting MAA-adduct formation. MTX is commonly used to treat RA and is being tested in CVD patients. MDA and AA are produced during lipidperoxidation and can interact to form MAA-adducts. MAA-adducts are found in atheromas and in diseased synovial tissue of RA patients. MTX scavenges the free radical O2− and prevents the formation of MAA-adducts. Scavenging O2− may be a mechanism by which MTX reduces inflammation and disease.
Collapse
Affiliation(s)
- Matthew C Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States
| | - Dahn L Clemens
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, United States
| | - Michael J Duryee
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States
| | - Cleofes Sarmiento
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States
| | - Andrew Chiou
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States
| | - Carlos D Hunter
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States
| | - Jun Tian
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States
| | - Lynell W Klassen
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, United States
| | - James R O'Dell
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, United States
| | - Geoffrey M Thiele
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, United States
| | - Ted R Mikuls
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States; Veterans Affairs (VA) Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, United States
| | - Daniel R Anderson
- Department of Internal Medicine, University of Nebraska Medical Center, 982650 Nebraska Medical Center, Omaha, NE 68198-2265, United States.
| |
Collapse
|